US20070270365A1 - Treatment of Eye Disorders Characterized by an Elevated Intraocular Pressure by siRNAs - Google Patents

Treatment of Eye Disorders Characterized by an Elevated Intraocular Pressure by siRNAs Download PDF

Info

Publication number
US20070270365A1
US20070270365A1 US11/574,169 US57416905A US2007270365A1 US 20070270365 A1 US20070270365 A1 US 20070270365A1 US 57416905 A US57416905 A US 57416905A US 2007270365 A1 US2007270365 A1 US 2007270365A1
Authority
US
United States
Prior art keywords
seq
sina
targeted
target gene
sequence selected
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/574,169
Other versions
US8030284B2 (en
Inventor
Ana Jimenez
Angela Sesto
Jose Roman
Irene Gascon
Gonzalo Gonzalez de Buitrago
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sylentis SA
Original Assignee
Sylentis SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=35967904&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20070270365(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from GBGB0418762.1A external-priority patent/GB0418762D0/en
Priority claimed from GB0503412A external-priority patent/GB0503412D0/en
Application filed by Sylentis SA filed Critical Sylentis SA
Assigned to SYLENTIS S.A.U. reassignment SYLENTIS S.A.U. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DE BUITRAGO, GONZALO GONZALEZ, SESTO, ANGELA, JIMENEZ, ANA I., GASCON, IRENE, ROMAN, JOSE P.
Publication of US20070270365A1 publication Critical patent/US20070270365A1/en
Application granted granted Critical
Publication of US8030284B2 publication Critical patent/US8030284B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Definitions

  • the present invention relates to methods and compositions for the treatment of eye disorders; in particular but not exclusively to the treatment of glaucoma.
  • the invention relates to the use of RNAI technology to downregulate the expression of aqueous formation genes and aqueous outflow genes. Methods and compositions for the treatment of eye disorders are also provided.
  • RNAI as a Tool to Downregulate Gene Expression
  • Gene targeting by homologous recombination is commonly used to determine gene function in mammals, but this is a costly and time-consuming process.
  • the functions of many genes can be determined after mRNA inhibition with ribozyme or antisense technologies, Although successful in some situations these technologies have been difficult to apply universally.
  • siRNA-directed “knockdown” has sparked a revolution In somatic cell genetics, allowing the inexpensive and rapid analysis of gene function in mammals.
  • dsRNA double-stranded RNA
  • the sequence of the first strand coincides with that of the corresponding region of the target messenger RNA (mRNA).
  • the second strand is complementary to this mRNA.
  • the resulting dsRNA turned out to be far more (several orders of magnitude) efficient than the corresponding single-stranded RNA molecules (in particular, antisense RNA).
  • Fire et al., 1998 named the phenomenon RNAi for RNA interference. This powerful gene silencing mechanism has been shown to operate in several species among most phylogenetic phyla.
  • RNAi begins when an enzyme named DICER encounters dsRNA and chops it into pieces called small-interfering RNAs or siRNAs.
  • This protein belongs to the RNase III nuclease family. A complex of proteins gathers up these RNA remains and uses their code as a guide to search out and destroy any RNAs in the cell with a matching sequence, such as target mRNA (for review see Bosher & Labouesse, 2000).
  • RNAi phenomenon (Akashi et al., 2001) might be summarized as follows:
  • Step 1 dsRNA recognition and scanning process.
  • Step 2 dsRNA cleavage through RNase III activity and production of siRNAs.
  • Step 3 association of the siRNAs and associated factors in RISC complexes.
  • Step 4 recognition of the complementary target mRNA.
  • Step 5 cleavage of the target mRNA in the centre of the region complementary to the siRNA.
  • Step 6 degradation of the target mRNA and recycling of the RISC complex.
  • RNAI RNAI-induced RNAI
  • mammalian cells have developed various protective phenomena against viral infections that could impede the use of this approach. Indeed, the presence of extremely low levels of viral dsRNA triggers an interferon response, resulting in a global non-specific suppression of translation, which in turn triggers apoptosis (Williams, 1997, Gil & Esteban, 2000).
  • RNAs folded in hairpin structures were used to inhibit the function of specific genes. This work was inspired by previous studies showing that some genes in Caenorhabditis elegans naturally regulate other genes through RNAi by coding for hairpin-structured RNAs. Tested in a variety of normal and cancer human and mouse cell lines, short hairpin RNAs (shRNAs) are able to silence genes as efficiently as their siRNA counterparts. Moreover, shRNAs exhibit better reassociation kinetics in vivo than equivalent duplexes.
  • RNAs small temporally regulated RNAs
  • stRNAs small temporally regulated RNAs
  • siRNAs In contrast with siRNAs, 22 nt long stRNAs downregulate expression of target mRNA after translational initiation without affecting mRNA integrity. Recent studies indicate that the two stRNAs first described in nematodes are the members of a huge family with hundreds of additional micro-RNAs (miRNAs) existing in metazoans (Grosshans & Slack, 2002).
  • RNAi is a broadly applicable method for gene silencing in vitro. Based on these results, RNAi has rapidly become a well recognized tool for validating (identifying and assigning) gene functions.
  • RNA interference employing short dsRNA oligonucleotides will, moreover, permit to decipher the function of genes being only partially sequenced. RNAi will therefore become inevitable in studies such as:
  • RNAi is a straight-forward tool to rapidly assess gene function and reveal null phenotypes.
  • RNAi may yield RNA-based drugs to treat human diseases.
  • Glaucoma is one of the leading causes of blindness, Approximately 15% of cases of blindness world-wide result from glaucoma. The most common type, primary open-angle glaucoma, has a prevalence of 1/200 in the general population over 40 years of age.
  • Glaucoma has been simply defined as the process of ocular tissue destruction caused by a sustained elevation of the Intra Ocular Pressure (IOP) above its normal physiological limits.
  • IOP Intra Ocular Pressure
  • glaucomas are characterised by an elevated IOP, although the level of elevation may vary.
  • the elevation is initially low (i.e., open angle glaucoma, melanocytic glaucoma) and some secondary glaucoma, retinal ganglion cell and optic nerve damage are slow to progress.
  • angle-closure glaucoma the sudden high rise in IOP often renders the eye blind, undoubtedly primarily due to a cessation of axoplasmic flow at the level of the lamina cribrosa.
  • tissue ischaemia has a part to play in the initiation or progression of the optic disc damage that occurs in glaucoma.
  • Retinal ganglion cell degeneration may be necrosis, but the possibility that it is apoptosis triggered by the rise in IOP is plausible, and the respective roles of nitric oxide and glutamate are thought to be relevant during progression of the disease (For a recent review on the subject see Osborne et al, 2003).
  • the absolute determinant in therapy selection is the amount of primary and/or induced change in pressure within the iridocorneal angle.
  • aqueous formation suppressors among them, carbonic anhydrase inhibitors, beta-adrenergic blocking agents, or alpha2-adrenoreceptor agonists
  • miotics i.e. parasympathomimetics—cholinergics-, or anticholinesterase inhibitors
  • uveoscleral outflow enhancers i.e. parasympathomimetics—cholinergics-, or anticholinesterase inhibitors
  • hyperosmotic agents that produce an osmotic pressure gradient across the blood/aqueous barrier within the ciliary epithelium. All four are used in the treatment of glaucoma, the first three commonly as emergency treatment and in long term control while the hyperosmotic agents are invaluable as emergency and preoperative treatment.
  • a fifth category of drugs, the neuroprotection agents, is beginning to emerge as an important possible addition to medical therapy. Indeed, observation that the NOS and glutamate levels are elevated in glaucoma and that they are involved in retinal ganglion cell necrosis or apoptosis has raised the possibility of neuroprotective therapies and even neuroregeneration.
  • NOS inhibitors, exciting amino acid antagonists, glutamate receptor antagonists, apoptosis inhibitors and calcium channel blockers are all potential candidates in the development of future glaucoma therapies.
  • the calcium channel blockers may reduce the effect of impaired microcirculation to the optic nerve head whilst potentially increasing outflow facility at the level of the trabecular cells.
  • the eye conditions may include glaucoma, uveitis, and inflammation.
  • the method is based on the downregulation of expression of genes involved in aqueous formation or aqueous outflow in the eye. Downregulation may be effected by the use of double stranded nucleic acid moieties, named siNA or small interfering NA that are directed at interfering with the mRNA expression of various candidate genes.
  • the siNA are preferably siRNA, although modified nucleic acids or similar chemically synthesised entities are also included within the scope of the invention.
  • Embodiments of the invention relate to topical application of siNA.
  • Embodiments of the invention also provide pharmaceutical compositions for use in the treatment of eye conditions.
  • the invention may be used within the fields of local eye treatments, of target genes involved in glaucoma pathogenesis, as well as the use of chemically synthesized entities to treat animal (including humans) diseases.
  • the present method is also suitable for the treatment of other diseases of the anterior chamber of the eye.
  • the method may be applied to the treatment of diseases characterised by altered aqueous formation or outflow in the eye.
  • diseases which may be treated include local conditions such as infections or inflammations, and general conditions such as uveitis or expression of systemic diseases.
  • certain embodiments of the invention provide treatment for diabetic retinopathy.
  • target genes whose expression levels may alter IOP. These genes can fall within the groups of genes involved in aqueous formation or the group of genes involved in aqueous outflow.
  • target genes whose expression levels may alter IOP.
  • Adrenergic Receptors beta1 and 2 and alpha 1A, 1B and 1D
  • ATPases alpha1, alpha2, alpha3, beta1, beta2
  • Endothelial Leukocyte Adhesion Molecule I (ELAM-1)
  • Angiotensin System Angiotensin II, Angiotensin II Converting Enzymes (ACE I and ACE II), Angiotensin II Receptors (ATR1 and ATR2) and Renin
  • RNAi Although the mechanisms for RNAi remain unknown, the steps required to generate the specific dsRNA oligonucleotides are clear. It has been shown that dsRNA duplex strands that are 21-26 nucleotides in length work most effectively in producing RNA interference. Selecting the right homologous region within the gene is also important. Factors such as the distance from start codon, the G/C content and the location of adenosine dimers are important when considering the generation of dsRNA for RNAi. One consequence of this, however, is that one may need to test several different sequences for the most efficient RNAi and this may become costly.
  • RNAi relies on the establishment of complex protein interactions, it is obvious that the mRNA target should be devoided of unrelated bound factors.
  • UTRs 5′ and 3′ untranslated regions
  • regions close to the start codon should be avoided as they may be richer in regulatory protein binding sites.
  • the sequence of the siRNA is therefore selected as follows:
  • each of the selected genes is introduced as a nucleotide sequence in a prediction program that takes into account all the variables described above for the design of optimal oligonucleotides.
  • This program scans any mRNA nucleotide sequence for regions susceptible to be targeted by siRNAs.
  • the output of this analysis is a score of possible siRNA oligonucleotides. The highest scores are used to design double stranded RNA oligonucleotides (typically 21 bp long, although other lengths are also possible) that are typically made by chemical synthesis.
  • modified nucleotides may also be used.
  • Affinity modified nucleosides as described in WO2005/044976 may be used.
  • This publication describes oligonucleotides comprising nucleosides modified so as to have increased or decreased affinity for their complementary nucleotide in the target mRNA and/or in the complementary siNA strand.
  • GB2406568 describes alternative modified oligonucleotides chemically modified to provide improved resistance to degradation or improved uptake.
  • modifications include phosphorothioate internucleotide linkages, 2′-O-methyl ribonucleotides, 2′-deoxy-fluoro ribonucleotides, 2′-deoxy ribonucleotides, “universal base” nucleotides, 5-C-methyl nucleotides, and inverted deoxyabasic residue incorporation.
  • WO2004/029212 describes oligonucleotides modified to enhance the stability of the siRNA or to increase targeting efficiency. Modifications include chemical cross linking between the two complementary strands of an siRNA and chemical modification of a 3′ terminus of a strand of an siRNA. Preferred modifications are internal modifications, for example, sugar modifications, nucleobase modifications and/or backbone modifications. 2′-fluoro modified ribonucleotides and 2′-deoxy ribonucleotides are described.
  • WO2005/040537 further recites modified oligonucleotides which may be used in the invention.
  • the present invention may use short hairpin NA (shNA); the two strands of the siNA molecule may be connected by a linker region, which may be a nucleotide linker or a non-nucleotide linker.
  • shNA short hairpin NA
  • siNA molecules may be used to target homologous regions.
  • WO2005/045037 describes the design of siNA molecules to target such homologous sequences, for example by incorporating non-canonical base pairs, for example mismatches and/or wobble base pairs, that can provide additional target sequences.
  • non-canonical base pairs for example, mismatches and/or wobble bases
  • non-canonical base pairs can be used to generate siNA molecules that target more than one gene sequence.
  • non-canonical base pairs such as UU and CC base pairs are used to generate siNA molecules that are capable of targeting sequences for differing targets that share sequence homology.
  • siNAs of the invention are designed to include nucleic acid sequence that is complementary to the nucleotide sequence that is conserved between homologous genes.
  • a single siNA can be used to inhibit expression of more than one gene instead of using more than one siNA molecule to target different genes.
  • siNA molecules of the invention are double stranded.
  • a siNA molecule of the invention may comprise blunt ends, that is, ends that do not include any overhanging nucleotides.
  • an siNA molecule of the invention can comprise one or more blunt ends.
  • the siNA molecules have 3′ overhangs.
  • siNA molecules of the invention may comprise duplex nucleic acid molecules with 3′ overhangs of n nucleotides (5 ⁇ n ⁇ 1). Elbashir (2001) shows that 21-nucleotide siRNA duplexes are most active when containing 3′-terminal dinucleotide overhangs.
  • Candidate oligonucleotides are further filtered for interspecies sequence conservation in order to facilitate the transition from animal to human clinical studies.
  • conserved oligonucleotides are used; this allows a single oligonucleotide sequence to be used in both animal models and human clinical trials.
  • GenBank Accession Numbers corresponding to our selected human target genes are shown in FIG. 1 .
  • alternative splicing produces a family of transcripts that differ in exon content.
  • the present invention allows individual targeting of each transcript form.
  • FIG. 2 Selected oligonucleotide sequences against which RNAi is directed are shown in FIG. 2 . Displayed sequences are the DNA sequences targeted by the siNA. Therefore, the invention would make use of NA duplexes with sequences complementary to the indicated DNA sequences.
  • target DNA need not necessarily be preceded by M or CA. Further, target DNA could be constituted by sequences included in FIG. 2 flanked by any contiguous sequence.
  • RNAs are preferably chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer. Substitution of one or both strands of a siRNA duplex by 2′-deoxy or 2′-O-methyl oligoribonucleotides abolished silencing in fly extract (Elbashir et al. 2001). In mammalian cells, however, it seems possible to substitute the sense siRNA by a 2′-O-methyl oligoribonucleotide (Ge et al. 2003).
  • siRNAs are obtained from commercial RNA oligo synthesis suppliers, which sell RNA-synthesis products of different quality and costs. In general, 21-nt RNAs are not too difficult to synthesize and are readily provided in a quality suitable for RNAi.
  • RNA synthesis reagents include Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Col., USA), Glen Research (Sterling, Va., USA), ChemGenes (Ashland, Mass., USA), and Cruachem (Glasgow, UK), Qiagen (Germany), Ambion (USA) and Invitrogen (Scotland).
  • the previous custom RNA synthesis companies are entitled to provide siRNAs with a license for target validation.
  • our siRNA suppliers are Ambion, Dharmacon and Invitrogen, companies that offer the traditional custom chemical synthesis service for siRNA, and supply the siRNA with HPLC purification and delivered in dry form along with RNase-free water.
  • a central web-based resource for RNAi and siRNA methodologies, along with links to additional siRNA related products and services, can be found on the website of above-mentioned suppliers.
  • RNA annealing step is necessary when working with single-stranded RNA molecules. It is critical that all handling steps be conducted under sterile, Rnase free conditions. To anneal the RNAs, the oligos must first be quantified by UV absorption at 260 nanometres (nm). The following protocol based on Elbashir et al. (2001) is then used for annealing:
  • each RNA oligo Separately aliquot and dilute each RNA oligo to a concentration of 50 ⁇ M.
  • RNA oligo solution Combine 30 ⁇ l of each RNA oligo solution and 15 ⁇ l of 5 ⁇ annealing buffer.
  • Final buffer concentration is: 100 mM potassium acetate, 30 mM HEPES-KOH pH 7.4, 2 mM magnesium acetate.
  • Final volume is 75 ⁇ l.
  • siRNA duplex Incubate the solution for 1 minute at 90° C., centrifuge the tube for 15 seconds, let sit for 1 hour at 37° C., then use at ambient temperature.
  • the solution can be stored frozen at ⁇ 20° C. and freeze-thawed up to 5 times.
  • the final concentration of siRNA duplex is usually 20 ⁇ M.
  • dsRNAs may be purchased from the suppliers.
  • Chemically modified nucleic acids may also be used.
  • an overview of the types of modification which may be used is given in WO03/070744, the contents of which are incorporated herein by reference. Particular attention is drawn to pages 11 to 21 of this publication. Other possible modifications are as described above. The skilled person will be aware of other types of chemical modification which may be incorporated into RNA molecules.
  • the cells used for these experiments were: rabbit non-pigmented ciliary epithelium cells NPE, human ciliary epithelium cells OMDC, and human embryonic kidney cells HEK293.
  • the cells are incubated with the corresponding siRNA duplexes, and analysis of the downregulation of expression of the target gene is carried out.
  • siRNA knockdown to specific phenotypes in cultured cells, it is necessary to demonstrate the reduction of targeted protein or at least demonstrate the reduction of the targeted mRNA.
  • mRNA levels of the target gene can be quantitated by real-time quantitative PCR (RT-PCR). Further, the protein levels can be determined in a variety of ways well known in the art, such as Western blot analysis with specific antibodies to the different target allow direct monitoring of the reduction of targeted protein.
  • RT-PCR real-time quantitative PCR
  • RNAiFect Transfection Reagent Qiagen
  • Lipofectamine 2000 Reagent Invitrogen
  • a typical transfection protocol can be performed as follows: For one well of a 6-well plate, we transfect using 100 nM as final concentration of siRNA. Following RNAiFect protocol, we seed, the day before transfection, 2-4 ⁇ 10 5 cells per well in 3 ml of an appropriate growth medium, containing DMEM, 10% serum, antibiotics and glutamine, and incubate cells under normal growth conditions (37° C. and 5% CO 2 ). On the day of transfection, cells have to be at 30-50% confluence. We dilute 15 ul of 20 uM siRNA duplex (corresponding to 100 nM final concentration) in 85 ul of Buffer EC-R, to give a final volume of 100 ul, and mix by vortexing.
  • RNAiFect Transfection Reagent for complex formation, we add 19 ul of RNAiFect Transfection Reagent to the diluted siRNA and mix by pipetting or vortexing. After incubating the samples for 10-15 minutes at room temperature to allow formation of transfection complexes, we add the complexes drop-wise onto the cells with 2.9 ml of fresh growth medium low in antibiotics. After swirling the plates to ensure uniform distribution of the transfection complexes, we incubate the cells under their normal growth conditions. The day after, the complexes are removed and fresh and complete growth medium is added. To monitor gene silencing, cells are collected at 24, 48 and 72 hours post-transfection. The Lipofectamine 2000 Reagent protocol is quite similar.
  • the diluted oligomer and the diluted Lipofectamine are combined to allow complex formation during a 20 minutes incubation at room temperature.
  • the efficiency of transfection may depend on the cell type, but also on the passage number and the confluency of the cells.
  • the time and the manner of formation of siRNA-liposome complexes are also critical. Low transfection efficiencies are the most frequent cause of unsuccessful silencing. Good transfection is a non-trivial issue and needs to be carefully examined for each new cell line to be used.
  • Transfection efficiency may be tested transfecting reporter genes, for example a CMV-driven EGFP-expression plasmid (e.g. from Clontech) or a B-Gal expression plasmid, and then assessed by phase contrast and/or fluorescence microscopy the next day.
  • a knock-down phenotype may become apparent after 1 to 3 days, or even later.
  • depletion of the protein may be observed by immunofluorescence or Western blotting.
  • RNA fractions extracted from cells were pre-treated with DNase I and used for reverse transcribed using a random primer.
  • PCR is amplified with a specific primer pair covering at least one exon-exon junction in order to control for amplification of pre-mRNAs.
  • RT/PCR of a non-targeted mRNA is also needed as control. Effective depletion of the mRNA yet undetectable reduction of target protein may indicate that a large reservoir of stable protein may exist in the cell.
  • RealTime PCR amplification can be used to test in a more precise way the mRNA decrease or disappearance.
  • Real-time reverse-transcriptase (RT) PCR quantitates the initial amount of the template most specifically, sensitively and reproducibly.
  • Real-time PCR monitors the fluorescence emitted during the reaction as an indicator of amplicon production during each PCR cycle. This signal increases in direct proportion to the amount of PCR product in a reaction. By recording the amount of fluorescence emission at each cycle, it is possible to monitor the PCR reaction during exponential phase where the first significant increase in the amount of PCR product correlates to the initial amount of target template.
  • qRT-PCR was performed according to the manufacturer protocol.
  • qRT-PCR quantitative RT-PCR
  • approximately 250 ng of total RNA were used for reverse transcription followed by PCR amplification with specific primers for each gene in reaction mixture containing Master SYBR Green I.
  • Basic PCR conditions comprised an initial step of 30 minutes at 91° C., followed by 40 cyles of 5 s at 95° C., 10 s at 62° C. and 15 s at 72° C. Specific primer sequences corresponding to each target gene were used. Quantification of b-actin mRNA was used as a control for data normalization.
  • Relative gene expression comparisons work best when the gene expression of the chosen endogenous/internal control is more abundant and remains constant, in proportion to total RNA, among the samples.
  • quantitation of an mRNA target can be normalised for differences in the amount of total RNA added to each reaction.
  • the present invention may comprise the administration of one or more species of siNA molecule simultaneously. These species may be selected to target one or more target genes.
  • siNA molecules of the Invention and formulations or compositions thereof may be administered directly or topically (e. g., locally) to the eye as is generally known In the art.
  • a siNA molecule can comprise a delivery vehicle, including liposomes, for administration to a subject.
  • Carriers and diluents and their salts can be present in pharmaceutically acceptable formulations.
  • Nucleic acid molecules can be administered to cells by a variety of methods known to those of skill in the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as biodegradable polymers, hydrogels, cyclodextrins poly (lactic-co-glycolic) acid (PLGA) and PLCA microspheres, biodegradable nanocapsules, and bioadhesive microspheres, or by proteinaceous vectors.
  • nucleic add molecules of the invention can also be formulated or complexed with polyethyleneimine and derivatives thereof, such as polyethyleneimine-polyethyleneglycol-N-acetylgalactosamine (PEI-PEG-GAL) or polyethyleneimine-polyethyleneglycol-tri-N-acetylgalactosamine (PEI-PEG-triGAL) derivatives.
  • polyethyleneimine-polyethyleneglycol-N-acetylgalactosamine PEI-PEG-GAL
  • PEI-PEG-triGAL polyethyleneimine-polyethyleneglycol-tri-N-acetylgalactosamine
  • a siNA molecule of the invention may be complexed with membrane disruptive agents and/or a cationic lipid or helper lipid molecule.
  • Delivery systems which may be used with the invention include, for example, aqueous and non aqueous gels, creams, multiple emulsions, microemulsions, liposomes, ointments, aqueous and non aqueous solutions, lotions, aerosols, hydrocarbon bases and powders, and can contain excipients such as solubilizers, permeation enhancers (e. g., fatty acids, fatty acid esters, fatty alcohols and amino acids), and hydrophilic polymers (e. g. , polycarbophil and polyvinylpyrolidone).
  • the pharmaceutically acceptable carrier is a liposome or a transdermal enhancer.
  • a pharmaceutical formulation of the invention is in a form suitable for administration, e.g., systemic or local administration, into a cell or subject, including for example a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, or by injection. Other factors are known in the art, and include considerations such as toxicity and forms that prevent the composition or formulation from exerting its effect.
  • compositions prepared for storage or administration that include a pharmaceutically effective amount of the desired compounds in a pharmaceutically acceptable carrier or diluent.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art.
  • preservatives, stabilizers, dyes and flavouring agents can be provided. These include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • antioxidants and suspending agents can be used.
  • a pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) of a disease state.
  • the pharmaceutically effective dose dependes on the type of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors that those skilled in the medical arts will recognize
  • an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered.
  • compositions of the invention can be administered in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and/or vehicles.
  • Formulations can be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavouring agents, colouring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients can be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets can be uncoated or they can be coated by known techniques. In some cases such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, caldum phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, caldum phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents can be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate
  • the aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more colouring agents, one or more flavouring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • colouring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavouring agents such as sucrose or saccharin.
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions can be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavouring agents can be added to provide palatable oral preparations. These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent exemplified by those already mentioned above.
  • Additional excipients for example sweetening, flavouring and colouring agents, can also be present.
  • compositions of the invention can also be in the form of oil-in-water emulsions.
  • the oily phase can be a vegetable oil or a mineral oil or mixtures of these.
  • Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions can also contain sweetening and flavouring agents.
  • Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can also contain a demulcent, a preservative and flavouring and colouring agents.
  • the pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension.
  • This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above.
  • a sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a non-toxic parentally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the nucleic acid molecules of the invention can also be administered in the form of suppositories, e. g., for rectal administration of the drug.
  • suppositories e. g., for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials include cocoa butter and polyethylene glycols.
  • Nucleic acid molecules of the invention can be administered parenterally in a sterile medium.
  • the drug depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
  • adjuvants such as local anaesthetics, preservatives and buffering agents can be dissolved in the vehicle.
  • the specific dose level for any particular subject depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
  • the composition can also be added to the animal feed or drinking water. It can be convenient to formulate the animal feed and drinking water compositions so that the animal takes in a therapeutically appropriate quantity of the composition along with its diet. It can also be convenient to present the composition as a premix for addition to the feed or drinking water.
  • nucleic acid molecules of the present invention can also be administered to a subject in combination with other therapeutic compounds to increase the overall therapeutic effect.
  • the use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects.
  • siNA molecules of the invention can be expressed within cells from eukaryotic promoters.
  • Recombinant vectors capable of expressing the siNA molecules can be delivered and persist in target cells.
  • vectors can be used that provide for transient expression of nucleic add molecules. Such vectors can be repeatedly administered as necessary.
  • the siNA molecule interacts with the target mRNA and generates an RNAi response. Delivery of siNA molecule expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from a subject followed by reintroduction into the subject, or by any other means that would allow for introduction into the desired target cell.
  • the New Zealand rabbit is the gold standard in experimental platforms designed to study IOP. It is easy to handle and it has a big eye, similar in size to the human organ. In addition, present equipment to measure IOP is not suited to use in animals with small eyes such as mice or rats. Finally, rabbits have an IOP (around 23 mm Hg) that can be brought down to up to 40% its value using local commercial hypotensive medication.
  • rabbit glaucoma models for example, surgically blocking episclerotic veins or artificially occluding the trabecular meshwork
  • normotensive rabbits since, in our hands, the pharmacological decrease in IOP can be easily and reproducibly measured.
  • the drugs were typically administered by instilling a small volume (typically 40 ⁇ L) on the corneal surface. Contralateral eyes were treated with the vehicle alone and could be used as controls in each experiment lest there is a sympathy phenomenon with the other eye. Multiple experiments in the same animal should be abolished.
  • IOP measurements were done using a contact tonometer (TONOPEN XL, Mentor, Norwell, Mass.).
  • the TonoPen tonometer is very convenient due to its reliability and small size. Measurements with this instrument were performed delicately applying the tonometer's sensor to the corneal surface.
  • rabbits were topically anesthetized (oxibuprocaine/tetracaine, 0.4%/1%, in a saline solution (1/4 v:v). The solution was applied (10 ⁇ l) to the cornea before each measurement of intraocular pressure was made. siRNA or saline was applied topically to the cornea in volumes of 40 ⁇ l.
  • the standard protocol for the siRNA application in rabbit was as follows. Doses of siRNA in saline solution (0.9% w/v) to a final volume of 40 ul, were applied to one eye each day during four consecutive days. The opposite eye was taken as a control and 40 ul of sterile saline (0.9% w/v) were instilled on it, at the same time points. The IOP was measured before each application and at 2 h, 4 h and 6 h following the instillation, during 10 days. Maximum responses were observed between second and third day. To compare the effect of siRNA with other hypotensive compounds, Xalatan (latanoprost 0.005%) and Trustop (Dorzolamide 2%) were assayed and IOP measured in the same conditions.
  • the first step was to perform experiments in cell cultures. For each target, several siRNAs were designed using a specific software according to the rules described before. Those with the best characteristics were selected to be tested. The siRNAs were applied to cell cultures, such as NPE, OMDC and HEK293. The effect of siRNAs over the target gene was analyzed by real time PCR and semi-quantitative PCR according to standard protocols. The gene target transcript levels were normalized using actin as housekeeping gene. Table I below shows representative results of real time PCR experiments for some of the target genes described previously. The values represent the mean of the percentage of siRNA interference over each gene expression once normalized with the control cells and their standard deviations.
  • the level of the different transcripts at both 24 and 48 h time points was significantly reduced after the siRNA treatment.
  • the siRNAs used in the Table correspond to the listed human siRNA targets given in FIG. 2 as follows.
  • siRNA3 rabbit sequence homologous to human SEQ. ID. 477 TABLE I siRNA treatment reduces the levels or target gene transcripts.
  • RNA was prepared from cells treated with different siRNAs for 24 and 48 h. The samples were analyzed by real time PCR using specific primers. The values show the mean expression levels of different transcripts normalized to actin relative to cell control.
  • FIG. 3 shows some representative semi-quantitative gels for some of the targets described above.
  • the decrease of the gene expression for each target gene depends on the efficiency in siRNA silencing.
  • the most effective siRNA obtained by in vitro studies was administered to the animal model.
  • RNA was prepared from cells treated with different siRNAs. The samples were analyzed by semi-quantitative PCR using specific primers.
  • the figure shows a representative semi-quantitative gel for Beta Adrenergic Receptor 2 expression (A) and other for Acetylcholinesterase expression (B).
  • A MW Marker
  • C Control cells
  • TC Transfection Control
  • 1 siRNA1
  • 2 siRNA2
  • 3 siRNA3
  • NC Negative Control.
  • the expression levels for each target depends on the efficiency in siRNA silencing.
  • the siRNAs used in the Figure correspond to the human targets given in FIG. 2 as follows:
  • siRNAs selected by the in vitro assays were applied to the animal model, following the protocol previously described. To avoid the effect of IOP fluctuations due to circadian cycles, all the applications were performed at the same time. To determine the siRNA effect, intraocular pressures (IOPs) were measured as previously mentioned.
  • ATP1B2 rabbit sequence identical to human SEQ. ID. 1820 TABLE II Effect of siRNAs on the reduction of IOP in normotensive New Zealand rabbit. The values represent the mean of the percentage of IOP reduction over the control (contralateral eye with vehicle alone) and their standard error (SEM).

Abstract

Sequences and protocols for treatment of eye conditions by use of RNA interference are disclosed. Target genes are selected from those responsible for aqueous flow or aqueous outflow, while particularly preferred conditions to be treated include glaucoma and uveitis.

Description

    FIELD OF THE INVENTION
  • The present invention relates to methods and compositions for the treatment of eye disorders; in particular but not exclusively to the treatment of glaucoma. In preferred embodiments, the invention relates to the use of RNAI technology to downregulate the expression of aqueous formation genes and aqueous outflow genes. Methods and compositions for the treatment of eye disorders are also provided.
  • BACKGROUND OF THE INVENTION
  • RNAI as a Tool to Downregulate Gene Expression
  • Gene targeting by homologous recombination is commonly used to determine gene function in mammals, but this is a costly and time-consuming process. Alternatively, the functions of many genes can be determined after mRNA inhibition with ribozyme or antisense technologies, Although successful in some situations these technologies have been difficult to apply universally. The advent of siRNA-directed “knockdown” has sparked a revolution In somatic cell genetics, allowing the inexpensive and rapid analysis of gene function in mammals.
  • Establishing a convenient and reliable method to knock-out gene expression at the mRNA level has been a recurrent theme in molecular biology over the last 15 years. In efforts to generate loss-of function cells or organisms, various molecules that included, for example, antisense sequences, ribozymes, and chimeric oligonucleotides have been tested, but the design of such molecules was based on trial and error, depending on the properties of the target gene.
  • Moreover, the desired effects were difficult to predict, and often only weak suppression achieved (Braasch & Corey, 2002).
  • After the discovery of the phenomenon in plants in the early 1990s, in 1998 Andy Fire and Craig Mello for the first time demonstrated with the worm Caenorhabditis elegans that dsRNA (double-stranded RNA) may specifically and selectively inhibit gene expression in an extremely efficient manner (Fire et al., 1998). In their experiment, the sequence of the first strand (the so-called sense RNA) coincides with that of the corresponding region of the target messenger RNA (mRNA). The second strand (antisense RNA) is complementary to this mRNA. The resulting dsRNA turned out to be far more (several orders of magnitude) efficient than the corresponding single-stranded RNA molecules (in particular, antisense RNA). Fire et al., 1998 named the phenomenon RNAi for RNA interference. This powerful gene silencing mechanism has been shown to operate in several species among most phylogenetic phyla.
  • RNAi begins when an enzyme named DICER encounters dsRNA and chops it into pieces called small-interfering RNAs or siRNAs. This protein belongs to the RNase III nuclease family. A complex of proteins gathers up these RNA remains and uses their code as a guide to search out and destroy any RNAs in the cell with a matching sequence, such as target mRNA (for review see Bosher & Labouesse, 2000).
  • The RNAi phenomenon (Akashi et al., 2001) might be summarized as follows:
  • Step 1: dsRNA recognition and scanning process.
  • Step 2: dsRNA cleavage through RNase III activity and production of siRNAs.
  • Step 3: association of the siRNAs and associated factors in RISC complexes.
  • Step 4: recognition of the complementary target mRNA.
  • Step 5: cleavage of the target mRNA in the centre of the region complementary to the siRNA.
  • Step 6: degradation of the target mRNA and recycling of the RISC complex.
  • In trying to apply the RNAI phenomenon as a technology for gene knockdown, it was soon realized that mammalian cells have developed various protective phenomena against viral infections that could impede the use of this approach. Indeed, the presence of extremely low levels of viral dsRNA triggers an interferon response, resulting in a global non-specific suppression of translation, which in turn triggers apoptosis (Williams, 1997, Gil & Esteban, 2000).
  • In 2000, a first attempt with dsRNA resulted in the specific inhibition of 3 genes (MmGFP under the control of the Elongation Factor 1a, E-cadherin, and c-mos) in the mouse oocyte and early embryo. Translational arrest, and thus a PKR response, was not observed as the embryos continued to develop (Wianny & Zernicka-Goetz, 2000). One year later, research at Ribopharma AG (Kulmbach, Germany) first demonstrated the functionality of RNAi in mammalian cells. Using short (20-24 base pairs) dsRNAs—which are called SIRPLEX™—they specifically switched off genes even in human cells without initiating the acute-phase response. Similar experiments carried out later by other research groups (Elbashir et al., 2001; Caplen et al., 2001) further confirmed these results.
  • A year later, Paddison et. al. (Paddison et al, 2002) tried to use small RNAs folded in hairpin structures to inhibit the function of specific genes. This work was inspired by previous studies showing that some genes in Caenorhabditis elegans naturally regulate other genes through RNAi by coding for hairpin-structured RNAs. Tested in a variety of normal and cancer human and mouse cell lines, short hairpin RNAs (shRNAs) are able to silence genes as efficiently as their siRNA counterparts. Moreover, shRNAs exhibit better reassociation kinetics in vivo than equivalent duplexes. Even more important, these authors generated transgenic cell lines engineered to synthesize shRNAs that exhibit a long-lasting suppressing effect throughout cell divisions (Eurogentec). Recently, another group of small RNAs (also comprised in the range of 21-25 nt) was shown to mediate downregulation of gene expression. These RNAs, known as small temporally regulated RNAs (stRNAs), have been described in Caenorhabditis elegans were they regulate timing of gene expression during development. It should be noted that stRNAs and siRNAs, despite obvious similarities, proceed through different modes of action (for review see Banedjee & Slack, 2002. In contrast with siRNAs, 22 nt long stRNAs downregulate expression of target mRNA after translational initiation without affecting mRNA integrity. Recent studies indicate that the two stRNAs first described in nematodes are the members of a huge family with hundreds of additional micro-RNAs (miRNAs) existing in metazoans (Grosshans & Slack, 2002).
  • Scientists have initially used RNAi in several systems, including Caenorhabditis elegans, Drosophilia, trypanosomes, and various other invertebrates. Moreover, using this approach, several groups have recently presented the specific suppression of protein biosynthesis in different mammalian cell lines—specifically in HeLa cells—showing that RNAi is a broadly applicable method for gene silencing in vitro. Based on these results, RNAi has rapidly become a well recognized tool for validating (identifying and assigning) gene functions. RNA interference employing short dsRNA oligonucleotides will, moreover, permit to decipher the function of genes being only partially sequenced. RNAi will therefore become inevitable in studies such as:
  • Inhibition of gene expression at the post-transcriptional level in eukaryotic cells. In this context, RNAi is a straight-forward tool to rapidly assess gene function and reveal null phenotypes.
  • Development of the RNAi technology for use in post-implantation embryos.
  • The predominant economic significance of RNA interference is established by its application as a therapeutic principle. As so, RNAi may yield RNA-based drugs to treat human diseases.
  • Glaucoma
  • Glaucoma is one of the leading causes of blindness, Approximately 15% of cases of blindness world-wide result from glaucoma. The most common type, primary open-angle glaucoma, has a prevalence of 1/200 in the general population over 40 years of age.
  • Glaucoma has been simply defined as the process of ocular tissue destruction caused by a sustained elevation of the Intra Ocular Pressure (IOP) above its normal physiological limits.
  • It is becoming increasingly clear that many forms of glaucoma have a genetic component, and much current research is focused on identifying chromosomal regions and genes that contribute to glaucoma. It is likely that the aetiology of OAG is multifactorial, resulting from a combination of mutations in more than one gene and as yet unidentified environmental factors. With regard to juvenile and adult-onset OAG, several loci have been identified. However, only one gene is known, namely the myocilin/TIGR (trabecular meshwork inducible glucocorticoid response) gene at the GLC1A locus on chromosome 1q21-q31. More than thirty mutations of this gene have been identified in ethnically diverse populations worldwide. Studies have shown that it is responsible for only about 5% of OAG overall (See reviews in Wirtz & Samples, 2003, and Khaw et al, 2004a).
  • Pathogenesis
  • Most glaucomas are characterised by an elevated IOP, although the level of elevation may vary. In those glaucomas in which the elevation is initially low (i.e., open angle glaucoma, melanocytic glaucoma) and some secondary glaucoma, retinal ganglion cell and optic nerve damage are slow to progress. In angle-closure glaucoma the sudden high rise in IOP often renders the eye blind, undoubtedly primarily due to a cessation of axoplasmic flow at the level of the lamina cribrosa.
  • In human studies, it has been widely accepted that tissue ischaemia has a part to play in the initiation or progression of the optic disc damage that occurs in glaucoma. Retinal ganglion cell degeneration may be necrosis, but the possibility that it is apoptosis triggered by the rise in IOP is plausible, and the respective roles of nitric oxide and glutamate are thought to be relevant during progression of the disease (For a recent review on the subject see Osborne et al, 2003).
  • Treatment
  • Although several aetiologies are involved in the glaucoma complex, the absolute determinant in therapy selection is the amount of primary and/or induced change in pressure within the iridocorneal angle.
  • Current therapies include medications or surgeries aimed at lowering this pressure, although the pathophysiological mechanisms by which elevated IOP leads to neuronal damage in glaucoma are unknown.
  • Medical suppression of an elevated IOP can be attempted using four types of drugs: the aqueous formation suppressors (among them, carbonic anhydrase inhibitors, beta-adrenergic blocking agents, or alpha2-adrenoreceptor agonists) miotics (i.e. parasympathomimetics—cholinergics-, or anticholinesterase inhibitors); uveoscleral outflow enhancers; and the hyperosmotic agents (that produce an osmotic pressure gradient across the blood/aqueous barrier within the ciliary epithelium). All four are used in the treatment of glaucoma, the first three commonly as emergency treatment and in long term control while the hyperosmotic agents are invaluable as emergency and preoperative treatment. A fifth category of drugs, the neuroprotection agents, is beginning to emerge as an important possible addition to medical therapy. Indeed, observation that the NOS and glutamate levels are elevated in glaucoma and that they are involved in retinal ganglion cell necrosis or apoptosis has raised the possibility of neuroprotective therapies and even neuroregeneration. Thus NOS inhibitors, exciting amino acid antagonists, glutamate receptor antagonists, apoptosis inhibitors and calcium channel blockers are all potential candidates in the development of future glaucoma therapies. The calcium channel blockers may reduce the effect of impaired microcirculation to the optic nerve head whilst potentially increasing outflow facility at the level of the trabecular cells.
  • Reviews of various eye disorders and their treatments are given in the references, in particular in Bunce (2005), Costagliola (1995, 2000), Cullinane (2002), Sakaguchi (2002), Shah (2000), and Wang (2005).
  • Currently our existing therapies must fall short of the mark and the practical difficulties associated with the assessment of outflow facility, the accurate monitoring of therapy and the complexity of surgical techniques all combine to confound the prognosis. The overriding factor in all glaucoma is the degeneration of the retinal ganglion cell, thus neuroprotection through effective ocular hypotension is the essential requirement of any therapy we utilise (for a recent review on the subject, see Khaw et al 2004b).
  • BRIEF SUMMARY OF THE INVENTION
  • In the present invention we describe a method for the treatment of eye conditions characterised by an altered IOP in animals, including humans. In particular, the eye conditions may Include glaucoma, uveitis, and inflammation. The method is based on the downregulation of expression of genes involved in aqueous formation or aqueous outflow in the eye. Downregulation may be effected by the use of double stranded nucleic acid moieties, named siNA or small interfering NA that are directed at interfering with the mRNA expression of various candidate genes. The siNA are preferably siRNA, although modified nucleic acids or similar chemically synthesised entities are also included within the scope of the invention.
  • Preferred embodiments of the invention relate to topical application of siNA. Embodiments of the invention also provide pharmaceutical compositions for use in the treatment of eye conditions. The invention may be used within the fields of local eye treatments, of target genes involved in glaucoma pathogenesis, as well as the use of chemically synthesized entities to treat animal (including humans) diseases.
  • In addition to the treatment of glaucoma, the present method is also suitable for the treatment of other diseases of the anterior chamber of the eye. In particular, the method may be applied to the treatment of diseases characterised by altered aqueous formation or outflow in the eye. Examples of conditions which may be treated include local conditions such as infections or inflammations, and general conditions such as uveitis or expression of systemic diseases. Further, certain embodiments of the invention provide treatment for diabetic retinopathy.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Target Genes
  • In the present invention, we define a list of target genes, whose expression levels may alter IOP. These genes can fall within the groups of genes involved in aqueous formation or the group of genes involved in aqueous outflow. Here is a list of our target genes:
  • Carbonic Anhydrases II, IV and XII
  • Adrenergic Receptors: beta1 and 2 and alpha 1A, 1B and 1D
  • Acetylcholinesterase
  • Cyclooxygenases 1 and 2
  • ATPases: alpha1, alpha2, alpha3, beta1, beta2
  • Endothelial Leukocyte Adhesion Molecule I (ELAM-1)
  • Angiotensin System: Angiotensin II, Angiotensin II Converting Enzymes (ACE I and ACE II), Angiotensin II Receptors (ATR1 and ATR2) and Renin
  • Cochlin
  • Design of siNA
  • Although the mechanisms for RNAi remain unknown, the steps required to generate the specific dsRNA oligonucleotides are clear. It has been shown that dsRNA duplex strands that are 21-26 nucleotides in length work most effectively in producing RNA interference. Selecting the right homologous region within the gene is also important. Factors such as the distance from start codon, the G/C content and the location of adenosine dimers are important when considering the generation of dsRNA for RNAi. One consequence of this, however, is that one may need to test several different sequences for the most efficient RNAi and this may become costly.
  • In 1999, Tuschl et al. deciphered the silencing effect of siRNAs showing that their efficiency is a function of the length of the duplex, the length of the 3′-end overhangs, and the sequence in these overhangs. Based on this founder work, Eurogentec recommends that the target mRNA region, and hence the sequence of the siRNA duplex, should be chosen using the following guidelines:
  • Since RNAi relies on the establishment of complex protein interactions, it is obvious that the mRNA target should be devoided of unrelated bound factors. In this context, both the 5′ and 3′ untranslated regions (UTRs) and regions close to the start codon should be avoided as they may be richer in regulatory protein binding sites. The sequence of the siRNA is therefore selected as follows:
      • In the mRNA sequence, a region located 50 to 100 nt downstream of the AUG start codon or upstream of stop codon is selected.
      • In this region, the following sequences are searched for: AA(N19), CA(N19).
      • The G/C percentage for each identified sequence is calculated. Ideally, the G/C content is 50% but it must less than 70% and greater than 30%.
      • Preferably, sequences containing following repetitions are avoided: AAA, CCC, GGG, TTT, AAAA, CCCC, GGGG, TTTT.
      • An accessibility prediction according to the secondary structure of the mRNA is carried out as well.
      • A BLAST is also performed (i.e. NCBI ESrs database) with the nucleotide sequence fitting best the previous criteria to ensure that only one gene will be inactivated.
  • In order to maximize the result's interpretation, the following precautions should be taken when using siRNAs:
      • Always test the sense and antisense single strands in separate experiments.
      • Try a scramble siRNA duplex. This should have the same nucleotide composition as your siRNA but lack significant sequence homology to any other gene (including yours).
      • If possible, knock-down the same gene with two independent siRNA duplexes to control the specificity of the silencing process.
  • Practically, each of the selected genes is introduced as a nucleotide sequence in a prediction program that takes into account all the variables described above for the design of optimal oligonucleotides. This program scans any mRNA nucleotide sequence for regions susceptible to be targeted by siRNAs. The output of this analysis is a score of possible siRNA oligonucleotides. The highest scores are used to design double stranded RNA oligonucleotides (typically 21 bp long, although other lengths are also possible) that are typically made by chemical synthesis.
  • In addition to siRNA, modified nucleotides may also be used. We plan to test several chemical modifications that are well known in the art. These modifications are aimed at increasing stability or availability of the siNA. Examples of suitable modifications are described in the publications referenced below, each of which is incorporated herein by reference.
  • Studies show that replacing the 3′-terminal nucleotide overhanging segments of a 21-mer siRNA duplex having two -nucleotide 3′-overhangs with deoxyribonucleotides does not have an adverse effect on RNAi activity. Replacing up to four nucleotides on each end of the siRNA with deoxyribonucleotides has been reported to be well tolerated, whereas complete substitution with deoxyribonucleotides results in no RNAI activity (Elbashir 2001). In addition, Elbashir et al. also report that substitution of siRNA with 2′-O-methyl nucleotides completely abolishes RNAi activity.
  • Affinity modified nucleosides as described in WO2005/044976 may be used. This publication describes oligonucleotides comprising nucleosides modified so as to have increased or decreased affinity for their complementary nucleotide in the target mRNA and/or in the complementary siNA strand.
  • GB2406568 describes alternative modified oligonucleotides chemically modified to provide improved resistance to degradation or improved uptake. Examples of such modifications include phosphorothioate internucleotide linkages, 2′-O-methyl ribonucleotides, 2′-deoxy-fluoro ribonucleotides, 2′-deoxy ribonucleotides, “universal base” nucleotides, 5-C-methyl nucleotides, and inverted deoxyabasic residue incorporation.
  • WO2004/029212 describes oligonucleotides modified to enhance the stability of the siRNA or to increase targeting efficiency. Modifications include chemical cross linking between the two complementary strands of an siRNA and chemical modification of a 3′ terminus of a strand of an siRNA. Preferred modifications are internal modifications, for example, sugar modifications, nucleobase modifications and/or backbone modifications. 2′-fluoro modified ribonucleotides and 2′-deoxy ribonucleotides are described.
  • WO2005/040537 further recites modified oligonucleotides which may be used in the invention.
  • As well as making use of dsNA and modified dsNA, the present invention may use short hairpin NA (shNA); the two strands of the siNA molecule may be connected by a linker region, which may be a nucleotide linker or a non-nucleotide linker.
  • In addition to siNA which is perfectly complementary to the target region, degenerate siNA sequences may be used to target homologous regions. WO2005/045037 describes the design of siNA molecules to target such homologous sequences, for example by incorporating non-canonical base pairs, for example mismatches and/or wobble base pairs, that can provide additional target sequences. In instances where mismatches are identified, non-canonical base pairs (for example, mismatches and/or wobble bases) can be used to generate siNA molecules that target more than one gene sequence. In a non-limiting example, non-canonical base pairs such as UU and CC base pairs are used to generate siNA molecules that are capable of targeting sequences for differing targets that share sequence homology. As such, one advantage of using siNAs of the invention is that a single siNA can be designed to include nucleic acid sequence that is complementary to the nucleotide sequence that is conserved between homologous genes. In this approach, a single siNA can be used to inhibit expression of more than one gene instead of using more than one siNA molecule to target different genes.
  • Preferred siNA molecules of the invention are double stranded. A siNA molecule of the invention may comprise blunt ends, that is, ends that do not include any overhanging nucleotides. In one embodiment, an siNA molecule of the invention can comprise one or more blunt ends. In preferred embodiments, the siNA molecules have 3′ overhangs. siNA molecules of the invention may comprise duplex nucleic acid molecules with 3′ overhangs of n nucleotides (5≧n≧1). Elbashir (2001) shows that 21-nucleotide siRNA duplexes are most active when containing 3′-terminal dinucleotide overhangs.
  • Candidate oligonucleotides are further filtered for interspecies sequence conservation in order to facilitate the transition from animal to human clinical studies. In preferred embodiments of the invention, conserved oligonucleotides are used; this allows a single oligonucleotide sequence to be used in both animal models and human clinical trials.
  • GenBank Accession Numbers corresponding to our selected human target genes are shown in FIG. 1. In some of these genes, alternative splicing produces a family of transcripts that differ in exon content. The present invention allows individual targeting of each transcript form.
  • Selected oligonucleotide sequences against which RNAi is directed are shown in FIG. 2. Displayed sequences are the DNA sequences targeted by the siNA. Therefore, the invention would make use of NA duplexes with sequences complementary to the indicated DNA sequences.
  • The sequences shown in FIG. 2 are not limiting. As a matter of fact, target DNA need not necessarily be preceded by M or CA. Further, target DNA could be constituted by sequences included in FIG. 2 flanked by any contiguous sequence.
  • In vitro and Animal Studies.
  • Obtaining siRNA Duplexes
  • RNAs are preferably chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer. Substitution of one or both strands of a siRNA duplex by 2′-deoxy or 2′-O-methyl oligoribonucleotides abolished silencing in fly extract (Elbashir et al. 2001). In mammalian cells, however, it seems possible to substitute the sense siRNA by a 2′-O-methyl oligoribonucleotide (Ge et al. 2003).
  • Most conveniently, siRNAs are obtained from commercial RNA oligo synthesis suppliers, which sell RNA-synthesis products of different quality and costs. In general, 21-nt RNAs are not too difficult to synthesize and are readily provided in a quality suitable for RNAi.
  • Suppliers of RNA synthesis reagents include Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Col., USA), Glen Research (Sterling, Va., USA), ChemGenes (Ashland, Mass., USA), and Cruachem (Glasgow, UK), Qiagen (Germany), Ambion (USA) and Invitrogen (Scotland). The previous custom RNA synthesis companies are entitled to provide siRNAs with a license for target validation. In particular, our siRNA suppliers are Ambion, Dharmacon and Invitrogen, companies that offer the traditional custom chemical synthesis service for siRNA, and supply the siRNA with HPLC purification and delivered in dry form along with RNase-free water. A central web-based resource for RNAi and siRNA methodologies, along with links to additional siRNA related products and services, can be found on the website of above-mentioned suppliers.
  • An annealing step is necessary when working with single-stranded RNA molecules. It is critical that all handling steps be conducted under sterile, Rnase free conditions. To anneal the RNAs, the oligos must first be quantified by UV absorption at 260 nanometres (nm). The following protocol based on Elbashir et al. (2001) is then used for annealing:
  • Separately aliquot and dilute each RNA oligo to a concentration of 50 μM.
  • Combine 30 μl of each RNA oligo solution and 15 μl of 5× annealing buffer. Final buffer concentration is: 100 mM potassium acetate, 30 mM HEPES-KOH pH 7.4, 2 mM magnesium acetate. Final volume is 75 μl.
  • Incubate the solution for 1 minute at 90° C., centrifuge the tube for 15 seconds, let sit for 1 hour at 37° C., then use at ambient temperature. The solution can be stored frozen at −20° C. and freeze-thawed up to 5 times. The final concentration of siRNA duplex is usually 20 μM.
  • Alternatively, already annealed dsRNAs may be purchased from the suppliers.
  • Chemically modified nucleic acids may also be used. For example, an overview of the types of modification which may be used is given in WO03/070744, the contents of which are incorporated herein by reference. Particular attention is drawn to pages 11 to 21 of this publication. Other possible modifications are as described above. The skilled person will be aware of other types of chemical modification which may be incorporated into RNA molecules.
  • In vitro System
  • To check the specificity of the siRNA interference different cell cultures that express the target genes were employed. The cells used for these experiments were: rabbit non-pigmented ciliary epithelium cells NPE, human ciliary epithelium cells OMDC, and human embryonic kidney cells HEK293. The cells are incubated with the corresponding siRNA duplexes, and analysis of the downregulation of expression of the target gene is carried out. For linking siRNA knockdown to specific phenotypes in cultured cells, it is necessary to demonstrate the reduction of targeted protein or at least demonstrate the reduction of the targeted mRNA.
  • mRNA levels of the target gene can be quantitated by real-time quantitative PCR (RT-PCR). Further, the protein levels can be determined in a variety of ways well known in the art, such as Western blot analysis with specific antibodies to the different target allow direct monitoring of the reduction of targeted protein.
  • Transfection of siRNA Duplexes
  • Several examples of techniques well known in the art are as follows: We can perform a single transfection of siRNA duplex using a cationic lipid, such as RNAiFect Transfection Reagent (Qiagen) and Lipofectamine 2000 Reagent (Invitrogen) and assay for silencing 24, 48 and 72 hours after transfection.
  • A typical transfection protocol can be performed as follows: For one well of a 6-well plate, we transfect using 100 nM as final concentration of siRNA. Following RNAiFect protocol, we seed, the day before transfection, 2-4×105 cells per well in 3 ml of an appropriate growth medium, containing DMEM, 10% serum, antibiotics and glutamine, and incubate cells under normal growth conditions (37° C. and 5% CO2). On the day of transfection, cells have to be at 30-50% confluence. We dilute 15 ul of 20 uM siRNA duplex (corresponding to 100 nM final concentration) in 85 ul of Buffer EC-R, to give a final volume of 100 ul, and mix by vortexing. For complex formation, we add 19 ul of RNAiFect Transfection Reagent to the diluted siRNA and mix by pipetting or vortexing. After incubating the samples for 10-15 minutes at room temperature to allow formation of transfection complexes, we add the complexes drop-wise onto the cells with 2.9 ml of fresh growth medium low in antibiotics. After swirling the plates to ensure uniform distribution of the transfection complexes, we incubate the cells under their normal growth conditions. The day after, the complexes are removed and fresh and complete growth medium is added. To monitor gene silencing, cells are collected at 24, 48 and 72 hours post-transfection. The Lipofectamine 2000 Reagent protocol is quite similar. The day before transfection, we seed 2-4×105 cells per well in 3 ml of an appropriate growth medium, containing DMEM, 10% serum, antibiotics and glutamine, and incubate cells under normal growth conditions (37° C. and 5% CO2). On the day of transfection, cells have to be at 30-50% confluence. We dilute 12.5 ul of 20 uM siRNA duplex (corresponding to 100 nM final concentration) in 250 ul of DMEM, to give a final volume of 262.5 ul, and mix. Also, 6 ul of Lipofectamine 2000 is diluted in 250 ul of DMEM and mixed. After a 5 minutes incubation at room temperature, the diluted oligomer and the diluted Lipofectamine are combined to allow complex formation during a 20 minutes incubation at room temperature. Afterwards, we add the complexes drop-wise onto the cells with 2 ml of fresh growth medium low in antibiotics and mix gently by rocking the plate back and forth, to ensure uniform distribution of the transfection complexes. We incubate the cells under their normal growth conditions and the day after, the complexes are removed and fresh and complete growth medium is added. To monitor gene silencing, cells are collected at 24, 48 and 72 hours post-transfection.
  • The efficiency of transfection may depend on the cell type, but also on the passage number and the confluency of the cells. The time and the manner of formation of siRNA-liposome complexes (e.g. inversion versus vortexing) are also critical. Low transfection efficiencies are the most frequent cause of unsuccessful silencing. Good transfection is a non-trivial issue and needs to be carefully examined for each new cell line to be used. Transfection efficiency may be tested transfecting reporter genes, for example a CMV-driven EGFP-expression plasmid (e.g. from Clontech) or a B-Gal expression plasmid, and then assessed by phase contrast and/or fluorescence microscopy the next day.
  • Testing of siRNA Duplexes
  • Depending on the abundance and the life time (or turnover) of the targeted protein, a knock-down phenotype may become apparent after 1 to 3 days, or even later. In cases where no phenotype is observed, depletion of the protein may be observed by immunofluorescence or Western blotting.
  • After transfections, total RNA fractions extracted from cells were pre-treated with DNase I and used for reverse transcribed using a random primer. PCR is amplified with a specific primer pair covering at least one exon-exon junction in order to control for amplification of pre-mRNAs. RT/PCR of a non-targeted mRNA is also needed as control. Effective depletion of the mRNA yet undetectable reduction of target protein may indicate that a large reservoir of stable protein may exist in the cell. Alternatively, RealTime PCR amplification can be used to test in a more precise way the mRNA decrease or disappearance. Real-time reverse-transcriptase (RT) PCR quantitates the initial amount of the template most specifically, sensitively and reproducibly. Real-time PCR monitors the fluorescence emitted during the reaction as an indicator of amplicon production during each PCR cycle. This signal increases in direct proportion to the amount of PCR product in a reaction. By recording the amount of fluorescence emission at each cycle, it is possible to monitor the PCR reaction during exponential phase where the first significant increase in the amount of PCR product correlates to the initial amount of target template.
  • To verify the interference pattern of the differentially expressing genes identified in the cell cultures, qRT-PCR was performed according to the manufacturer protocol. For quantitative RT-PCR (qRT-PCR), approximately 250 ng of total RNA were used for reverse transcription followed by PCR amplification with specific primers for each gene in reaction mixture containing Master SYBR Green I. Basic PCR conditions comprised an initial step of 30 minutes at 91° C., followed by 40 cyles of 5 s at 95° C., 10 s at 62° C. and 15 s at 72° C. Specific primer sequences corresponding to each target gene were used. Quantification of b-actin mRNA was used as a control for data normalization. Relative gene expression comparisons work best when the gene expression of the chosen endogenous/internal control is more abundant and remains constant, in proportion to total RNA, among the samples. By using an invariant endogenous control as an active reference, quantitation of an mRNA target can be normalised for differences in the amount of total RNA added to each reaction.
  • Pharmaceutical Formulations
  • The present invention may comprise the administration of one or more species of siNA molecule simultaneously. These species may be selected to target one or more target genes.
  • The siNA molecules of the Invention and formulations or compositions thereof may be administered directly or topically (e. g., locally) to the eye as is generally known In the art. For example, a siNA molecule can comprise a delivery vehicle, including liposomes, for administration to a subject. Carriers and diluents and their salts can be present in pharmaceutically acceptable formulations. Nucleic acid molecules can be administered to cells by a variety of methods known to those of skill in the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as biodegradable polymers, hydrogels, cyclodextrins poly (lactic-co-glycolic) acid (PLGA) and PLCA microspheres, biodegradable nanocapsules, and bioadhesive microspheres, or by proteinaceous vectors. In another embodiment, the nucleic add molecules of the invention can also be formulated or complexed with polyethyleneimine and derivatives thereof, such as polyethyleneimine-polyethyleneglycol-N-acetylgalactosamine (PEI-PEG-GAL) or polyethyleneimine-polyethyleneglycol-tri-N-acetylgalactosamine (PEI-PEG-triGAL) derivatives.
  • A siNA molecule of the invention may be complexed with membrane disruptive agents and/or a cationic lipid or helper lipid molecule.
  • Delivery systems which may be used with the invention include, for example, aqueous and non aqueous gels, creams, multiple emulsions, microemulsions, liposomes, ointments, aqueous and non aqueous solutions, lotions, aerosols, hydrocarbon bases and powders, and can contain excipients such as solubilizers, permeation enhancers (e. g., fatty acids, fatty acid esters, fatty alcohols and amino acids), and hydrophilic polymers (e. g. , polycarbophil and polyvinylpyrolidone). In one embodiment, the pharmaceutically acceptable carrier is a liposome or a transdermal enhancer.
  • A pharmaceutical formulation of the invention is in a form suitable for administration, e.g., systemic or local administration, into a cell or subject, including for example a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, or by injection. Other factors are known in the art, and include considerations such as toxicity and forms that prevent the composition or formulation from exerting its effect.
  • The present invention also includes compositions prepared for storage or administration that include a pharmaceutically effective amount of the desired compounds in a pharmaceutically acceptable carrier or diluent. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art. For example, preservatives, stabilizers, dyes and flavouring agents can be provided. These include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. In addition, antioxidants and suspending agents can be used.
  • A pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) of a disease state. The pharmaceutically effective dose dependes on the type of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors that those skilled in the medical arts will recognize
  • Generally, an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered.
  • The formulations of the invention can be administered in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and/or vehicles. Formulations can be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavouring agents, colouring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • These excipients can be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets can be uncoated or they can be coated by known techniques. In some cases such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, caldum phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents can be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more colouring agents, one or more flavouring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • Oily suspensions can be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavouring agents can be added to provide palatable oral preparations. These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents or suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavouring and colouring agents, can also be present.
  • Pharmaceutical compositions of the invention can also be in the form of oil-in-water emulsions. The oily phase can be a vegetable oil or a mineral oil or mixtures of these. Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions can also contain sweetening and flavouring agents.
  • Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can also contain a demulcent, a preservative and flavouring and colouring agents. The pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension.
  • This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above.
  • A sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
  • The nucleic acid molecules of the invention can also be administered in the form of suppositories, e. g., for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols.
  • Nucleic acid molecules of the invention can be administered parenterally in a sterile medium. The drug, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anaesthetics, preservatives and buffering agents can be dissolved in the vehicle.
  • It is understood that the specific dose level for any particular subject depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
  • For administration to non-human animals, the composition can also be added to the animal feed or drinking water. It can be convenient to formulate the animal feed and drinking water compositions so that the animal takes in a therapeutically appropriate quantity of the composition along with its diet. It can also be convenient to present the composition as a premix for addition to the feed or drinking water.
  • The nucleic acid molecules of the present invention can also be administered to a subject in combination with other therapeutic compounds to increase the overall therapeutic effect. The use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects.
  • Alternatively, certain siNA molecules of the invention can be expressed within cells from eukaryotic promoters. Recombinant vectors capable of expressing the siNA molecules can be delivered and persist in target cells. Alternatively, vectors can be used that provide for transient expression of nucleic add molecules. Such vectors can be repeatedly administered as necessary. Once expressed, the siNA molecule interacts with the target mRNA and generates an RNAi response. Delivery of siNA molecule expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from a subject followed by reintroduction into the subject, or by any other means that would allow for introduction into the desired target cell.
  • Animal Studies
  • The New Zealand rabbit is the gold standard in experimental platforms designed to study IOP. It is easy to handle and it has a big eye, similar in size to the human organ. In addition, present equipment to measure IOP is not suited to use in animals with small eyes such as mice or rats. Finally, rabbits have an IOP (around 23 mm Hg) that can be brought down to up to 40% its value using local commercial hypotensive medication. Thus, although it is possible to generate rabbit glaucoma models (for example, surgically blocking episclerotic veins or artificially occluding the trabecular meshwork), we have used normotensive rabbits since, in our hands, the pharmacological decrease in IOP can be easily and reproducibly measured.
  • Experimental Protocol
  • Normotensive New Zealand White rabbits (males, 2-3 kg) were used. The animals were kept in individual cages with free access to food and water. They were submitted to artificial 12 hours light/darkness cycles, to avoid uncontrolled circadian oscillations of IOP Animal handling and treatment were carried out in accordance with the European Communities Council Directive (86/609/EEC) and the statement of the Association for Research in Vision and Ophthalmology on the Use of Animals in Ophthalmic and Vision Research.
  • The drugs were typically administered by instilling a small volume (typically 40 μL) on the corneal surface. Contralateral eyes were treated with the vehicle alone and could be used as controls in each experiment lest there is a sympathy phenomenon with the other eye. Multiple experiments in the same animal should be abolished.
  • IOP measurements were done using a contact tonometer (TONOPEN XL, Mentor, Norwell, Mass.). The TonoPen tonometer is very convenient due to its reliability and small size. Measurements with this instrument were performed delicately applying the tonometer's sensor to the corneal surface. This device has been shown to be the tonometer of choice for measuring intraocular pressures within the range of 3 to 30 mm Hg in rabbits (Abrams et al., 1996). All measurements fell within this interval: the mean baseline value of intraocular pressure was 17.0±0.39 mm Hg (n=100). Because IOP changes from the night to day, all the experiments were performed at the same time to allow IOP more stable and permit an objective comparison with vehicle treatment. In order to avoid distress to the animal, rabbits were topically anesthetized (oxibuprocaine/tetracaine, 0.4%/1%, in a saline solution (1/4 v:v). The solution was applied (10 μl) to the cornea before each measurement of intraocular pressure was made. siRNA or saline was applied topically to the cornea in volumes of 40 μl.
  • The standard protocol for the siRNA application in rabbit was as follows. Doses of siRNA in saline solution (0.9% w/v) to a final volume of 40 ul, were applied to one eye each day during four consecutive days. The opposite eye was taken as a control and 40 ul of sterile saline (0.9% w/v) were instilled on it, at the same time points. The IOP was measured before each application and at 2 h, 4 h and 6 h following the instillation, during 10 days. Maximum responses were observed between second and third day. To compare the effect of siRNA with other hypotensive compounds, Xalatan (latanoprost 0.005%) and Trustop (Dorzolamide 2%) were assayed and IOP measured in the same conditions.
  • Results
  • EXAMPLE 1 In vitro Assays
  • To determine the inhibition of the different targets involved in glaucoma using RNAi technology, the first step was to perform experiments in cell cultures. For each target, several siRNAs were designed using a specific software according to the rules described before. Those with the best characteristics were selected to be tested. The siRNAs were applied to cell cultures, such as NPE, OMDC and HEK293. The effect of siRNAs over the target gene was analyzed by real time PCR and semi-quantitative PCR according to standard protocols. The gene target transcript levels were normalized using actin as housekeeping gene. Table I below shows representative results of real time PCR experiments for some of the target genes described previously. The values represent the mean of the percentage of siRNA interference over each gene expression once normalized with the control cells and their standard deviations. Compared to the control cells, the level of the different transcripts at both 24 and 48 h time points was significantly reduced after the siRNA treatment. In the Table are included some of the different siRNAs that were tested and their different efficacies in the interference of the target gene. The siRNAs used in the Table correspond to the listed human siRNA targets given in FIG. 2 as follows.
  • AC2:
      • siRNA1: rabbit sequence homologous to human SEQ. ID. 73
      • siRNA2: rabbit sequence identical to human SEQ. ID. 54
      • siRNA3: rabbit sequence identical to human SEQ. ID. 66
  • PTGS1
      • siRNA1: rabbit sequence homologous to human SEQ. ID. 353
      • siRNA2: rabbit sequence homologous to human SEQ. ID. 369
  • PTGS2
      • siRNA1: rabbit sequence identical to human SEQ. ID. 426
      • siRNA2: rabbit sequence homologous to human SEQ. ID. 421
  • siRNA3: rabbit sequence homologous to human SEQ. ID. 477
    TABLE I
    siRNA treatment reduces the levels or target gene transcripts. RNA
    was prepared from cells treated with different siRNAs for 24 and
    48 h. The samples were analyzed by real time PCR using specific
    primers. The values show the mean expression levels of different
    transcripts normalized to actin relative to cell control.
    % of gene transcript level
    Target 24 h 48 h
    AC2 siRNA1 76.25 ± 12.60 84.57 ± 14.70
    siRNA2 37.97 ± 9.78  61.45 ± 9.62 
    siRNA3 35.30 ± 9.73  51.14 ± 16.49
    PTGS1 siRNA1 42.25 ± 13.76 42.68 ± 17.00
    siRNA2 34.98 ± 14.33 26.30 ± 10.91
    PTGS2 siRNA1 68.68 ± 12.48 70.17 ± 19.21
    siRNA2 81.00 ± 13.54 66.85 ± 18.67
    siRNA3 75.45 ± 14.71 61.83 ± 16.96
  • FIG. 3 shows some representative semi-quantitative gels for some of the targets described above. The decrease of the gene expression for each target gene depends on the efficiency in siRNA silencing. For each target, the most effective siRNA obtained by in vitro studies was administered to the animal model. RNA was prepared from cells treated with different siRNAs. The samples were analyzed by semi-quantitative PCR using specific primers. The figure shows a representative semi-quantitative gel for Beta Adrenergic Receptor 2 expression (A) and other for Acetylcholinesterase expression (B). M: MW Marker; C: Control cells; TC: Transfection Control; 1: siRNA1; 2: siRNA2; 3: siRNA3; NC: Negative Control. The expression levels for each target depends on the efficiency in siRNA silencing. The siRNAs used in the Figure correspond to the human targets given in FIG. 2 as follows:
  • Panel A (Beta Adrenergic Receptor 2)
      • 1: rabbit sequence homologous to human SEQ. ID. 122
      • 2: rabbit sequence identical to human SEQ. ID. 125
      • 3: rabbit sequence homologous to human SEQ. ID. 139
  • Panel B (Acetyicholinesterase)
      • 1: rabbit sequence homologous to human SEQ. ID. 162
      • 2: rabbit sequence homologous to human SEQ. ID. 177
    EXAMPLE 2 In vivo Assays
  • Previously to the siRNA therapeutical application, the in vivo assays were validated to determine the proper siRNA delivery.
  • Those siRNAs selected by the in vitro assays were applied to the animal model, following the protocol previously described. To avoid the effect of IOP fluctuations due to circadian cycles, all the applications were performed at the same time. To determine the siRNA effect, intraocular pressures (IOPs) were measured as previously mentioned.
  • Since glaucoma pathology presents an increase in the intraocular pressure, the aim was to obtain a decrease in its levels following siRNA application.
  • Most of the results for the different targets showed a significant decrease in IOP levels comparing with controls and also with commercial drugs (Latanoprost and Dorzolamide) and the animals treated with vehicle alone (negative control) didn't present any significant change in their IOP baseline. The data are summarized in table II where values represent the mean of the maximum percentage of IOP reduction after siRNA treatment once normalized and their standard deviations. The decrease in IOP was statistically significant for all the treated targets. These results indicate that siRNAs and commercial drugs act in a similar way, reducing IOP levels around 20%, although siRNAs present a more maintained effect. No secondary effects were observed in the animals along the experimental protocols. The siRNAs used in these experiments correspond to the human targets given in FIG. 2 as follows:
      • AC2: rabbit sequence homologous to human SEQ. ID. 73
      • AC4: rabbit sequence identical to human SEQ. ID. 5
      • AC12: rabbit sequence identical to human SEQ. ID. 522
      • ADRB1: rabbit sequence identical to human SEQ. ID. 105
      • ADRB2: rabbit sequence homologous to human SEQ. ID. 139
      • ADRA1A: rabbit sequence homologous to human SEQ. ID. 546
      • ADRA1B: rabbit sequence homologous to human SEQ. ID. 619
      • ACHE: rabbit sequence homologous to human SEQ. ID. 189
      • PTGS1: rabbit sequence homologous to human SEQ. ID. 322
      • PTGS2: rabbit sequence identical to human SEQ. ID. 426
      • SELE: rabbit sequence homologous to human SEQ. ID. 262
      • ACE1: rabbit sequence homologous to human SEQ. ID. 866
      • 10 AGTR1: rabbit sequence homologous to human SEQ. ID. 705
      • AGTR2: rabbit sequence identical to human SEQ. ID. 774
      • ATP1A1: rabbit sequence identical to human SEQ. ID. 1399
  • ATP1B2: rabbit sequence identical to human SEQ. ID. 1820
    TABLE II
    Effect of siRNAs on the reduction of IOP in normotensive New
    Zealand rabbit. The values represent the mean of the percentage
    of IOP reduction over the control (contralateral eye with
    vehicle alone) and their standard error (SEM).
    Target IOP reduction (% of the control)
    AC2 24.84 ± 3.41
    AC4 14.47 ± 5.00
    AC12 24.30 ± 1.29
    ADRB1 28.04 ± 2.98
    ADRB2 21.18 ± 1.88
    ADRA1A  9.51 ± 1.04
    ADRA1B 17.48 ± 1.30
    ACHE 25.25 ± 2.70
    PTGS1 14.62 ± 1.93
    PTGS2 23.78 ± 2.27
    SELE 21.80 ± 1.74
    ACE1 17.51 ± 1.28
    AGTR1  9.72 ± 1.35
    AGTR2 11.22 ± 1.53
    ATP1A1 18.13 ± 1.39
    ATP1B2 16.32 ± 0.91
    Latanoprost 25.46 ± 5.24
    Dorzolamide 16.41 ± 2.38
  • REFERENCES
    • Abrams L S, Vitale S, Jampel H D. Comparison of three tonometers for measuring intraocular pressure in rabbits. Invest Ophthalmol Vis Sci. 1996 April;37(5):940-4.
    • Akashi H, Miyagishi M, Taira K. Suppression of gene expression by RNA interference in cultured plant cells. Antisense Nucleic Acid Drug Dev, 2001, 11(6):359-67.
    • Banerjee D, Slack F. Control of developmental timing by small temporal RNAs: a paradigm for RNA-mediated regulation of gene expression. Bioessays, 2002, 24(2):119-29.
    • Bhattacharya S K, Annangudi S P, Salomon R G, Kuchtey R W, Peachey N S, Crabb J W. Cochlin deposits in the trabecular meshwork of the glaucomatous DBA/2J mouse. Exp Eye Res. 2005a May;80(5):741-4.
    • Bhattacharya S K, Rockwood E J, Smith S D, Bonilha V L, Crabb J S, Kuchtey R W, Robertson N G, Peachey N S, Morton C C, Crabb J W. Proteomics reveal Cochlin deposits associated with glaucomatous trabecular meshwork. J Biol Chem. 2005b Feb. 18;280(7):6080-4. Epub 2004 Dec. 3.
    • Bosher J M, Labouesse M. RNA interference: genetic wand and genetic watchdog. Nat Cell Biol, 2000, 2(2):E31-6.
    • Braasch D A, Corey D R. Novel antisense and peptide nucleic acid strategies for controlling gene expression. Biochemistry, 2002, 41(14):4503-10
    • Bunce C, Hitchings R A, Van Duijn C M, De Jong P T, Vingerling J R. Associations between the deletion polymorphism of the angiotensin 1-converting enzyme gene and ocular signs of primary open-angle glaucoma. Graefes Arch Clin Exp Ophthalmol. 2005 April; 243(4):294-9. Epub 2004 Oct. 13.
    • Caplen, N. J., Parrish, S., Imani, F., Fire, A. & Morgan, R. A. Specific inhibition of gene expression by small double stranded RNAs in invertebrate and vertebrate systems. Proc. Natl. Acad. Sci. USA, 2001, 98: 9742-9747.
    • Costagliola C, Verolino M, De Rosa M L, Iaccarino G, Ciancaglini M, Mastropasqua L. Effect of oral iosartan potassium administration on intraocular pressure in normotensive and glaucomatous human subjects. Exp Eye Res. 2000 August;71(2):167-71.
    • Costagliola C, Di Benedetto R. De Caprio L, Verde R, Mastropasqua L. Effect of oral captopril (SQ 14225) on intraocular pressure in man. Eur J Ophthalmol. 1995 January-March;5(1):19-25.
    • Cullinane A B, Leung P S, Ortego J, Coca-Prados M, Harvey B J. Renin-angiotensin system expression and secretory function in cultured human ciliary body non-pigmented epithelium. Br J Ophthalmol. 2002 June;86(6):676-83.
    • Elbashir S M, Lendeckel W, Tuschl T. RNA interference is mediated by 21- and 22-nucleotide RNAS. Genes Dev, 2001, 15(2):188-200.
    • Fire A, Xu S, Montgomery M K, Kostas S A, Driver S E, Mello C C. Potent and specific genetic interference by double stranded RNA in Caenorhabditis elegans. Nature, 1998, 391(6669):806-11.
    • Ge Q, McManus M T, Nguyen T, Shen C H, Sharp P A, Eisen H N, Chen J. RNA interference of influenza virus production by directly targeting mRNA for degradation and indirectly inhibiting all viral RNA transcription. Proc Natl Acad Sci USA., 2003; 100(5):2718-23.
    • Gil J, Esteban M. Induction of apoptosis by the dsRNA-dependent protein kinase (PKR): mechanism of action. Apoptosis, 2000, 5(2):107-14.
    • Grosshans H, Slack F J. Micro-RNAs: small is plentiful. J Cell Biol, 2002, 156(1):17-21.
    • Hara H, Ichikawa M, Oku H, Shimazawa M, Araie M. Bunazosin, a selective alpha1-adrenoceptor antagonist, as an anti-glaucoma drug: effects on ocular circulation and retinal neuronal damage. Cardiovasc Drug Rev. 2005 Spring;23(1):43-56.
    • Khaw P T, Shah P, Elkington A R. Glaucoma-1: diagnosis. BMJ, 2004a, 328:97-9.
    • Khaw P T, Shah P, Elkington A R. Glaucoma-2: treatment. BMJ, 2004b, 328:156-8.
    • Osborne N N, Chidlow G, Wood J, Casson R. Some current ideas on the pathogenesis and the role of neuroprotection in glaucomatous optic neuropathy. Eur J Ophthalmol. 2003 April; 13 Suppl 3:S19-26.
    • Paddison P J, Caudy A A, Bernstein E, Hannon G J, Conklin D S. Short hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian cells. Genes Dev, 2002, 16(8):948-58.
    • Sakaguchi H, Takai S, Sakaguchi M, Sugiyama T, Ishihara T, Yao Y, Miyazaki M, Ikeda T. Chymase and angiotensin converting enzyme activities in a hamster model of glaucoma filtering surgery. Curr Eye Res, 2002 May;24(5):325-31.
    • Shah G B, Sharma S, Mehta A A, Goyal R K. Oculohypotensive effect of angiotensin-converting enzyme inhibitors in acute and chronic models of glaucoma. J Cardiovasc Pharmacol. 2000 August;36(2):169-75.
    • Tuschl T, Zamore P D, Lehmann R, Bartel D P, Sharp P A. Targeted mRNA degradation by double-stranded RNA in vitro. Genes Dev., 1999; 13(24):3191-7.
    • Wang R F, Podos S M, Mittag T W, Yokoyoma T. Effect of CS-088, an angiotensin AT1 receptor antagonist, on intraocular pressure in glaucomatous monkey eyes. Exp Eye Res. 2005 May;80(5):629-32. Epub 2005 Jan. 4.
    • Wianny F, Zernicka-Goetz M. Specific interference with gene function by double-stranded RNA in early mouse development. Nat Cell Biol, 2000, 2(2):70-5.
    • Williams B R. Role of the double-stranded RNA-activated protein kinase (PKR) in cell regulation. Biochem Soc Trans, 1997, 25(2):509-13.
    • Wirtz M K, Samples J R. The genetic loci of open-angle glaucoma. Ophthalmol Clin North Am. 2003 16:505-14

Claims (53)

1. Use of siNA in the preparation of a medicament for use in a method of treatment of an eye condition, comprising downregulating expression of a target gene in a patient in need of treatment, the gene being selected from the group comprising aqueous formation genes and aqueous outflow genes.
2. The use of claim 1, wherein the eye condition is characterised by an altered intra-ocular pressure (IOP) in the patient.
3. The use of claim 1 or 2, wherein the eye condition is selected from the group comprising glaucoma, infection, inflammation, uveitis, and expression of systemic diseases.
4. The use of any preceding claim, wherein the eye condition is glaucoma.
5. The use of any preceding claim, wherein the eye condition is diabetic retinopathy.
6. The use of any preceding claim, wherein the target gene expression is downregulated in the eye of the patient.
7. The use of any preceding claim, wherein the target gene is selected from the group comprising: Carbonic Anhydrases II, IV and XII; Adrenergic Receptors: beta1 and 2 and alpha 1A, 1Band 1D; Acetylcholinesterase; Cyclooxygenases 1 and 2; ATPases: alpha1, alpha2, alpha3, beta1, beta2; Endothelial Leukocyte Adhesion Molecule I (ELAM-1); Angiotensin System: Angiotensin II, Angiotensin II Converting Enzymes (ACE I and ACE II), Angiotensin II Receptors (ATR1 and ATR2) and Renin; Cochlin.
8. The use of any preceding claim wherein the siNA is siRNA.
9. The use of claim 8 wherein the siRNA is dsRNA.
10. The use of claim 8 wherein the siRNA is shRNA.
11. The use of any preceding claim wherein the siNA comprises a modified oligonucleotide.
12. The use of any preceding claim wherein the siNA is administered topically to the eye of a patient.
13. The use of claim 12, wherein the siNA is administered to the cornea of a patient.
14. The use of any preceding claim wherein a plurality of species of siNA are used.
15. The use of claim 14 wherein said plurality of species are targeted to the same mRNA species.
16. The use of claim 14 wherein said plurality of species are targeted to different mRNA species.
17. The use of any preceding claim wherein the siNA is targeted to a sequence selected from SEQ ID 1 to SEQ ID 1829.
18. The use of claim 17 wherein the target gene is carbonic anhydrase IV, and the siNA is targeted to a sequence selected from SEQ ID 1 to SEQ ID 46.
19. The use of claim 17 wherein the target gene is carbonic anhydrase II, and the siNA is targeted to a sequence selected from SEQ ID 47 to SEQ ID 98.
20. The use of claim 17 wherein the target gene is beta adrenergic receptor 1, and the siNA is targeted to a sequence selected from SEQ ID 99 to SEQ ID 109.
21. The use of claim 17 wherein the target gene is beta adrenergic receptor 2, and the siNA is targeted to a sequence selected from SEQ ID 110 to SEQ ID 160.
22. The use of claim 17 wherein the target gene is acetylcholinesterase, and the siNA is targeted to a sequence selected from SEQ ID 161 to SEQ ID 190.
23. The use of claim 17 wherein the target gene is ELAM-1 (selectin E), and the siNA is targeted to a sequence selected from SEQ ID 191 to SEQ ID 318.
24. The use of claim 17 wherein the target gene is prostaglandin endoperoxide synthase 1, and the siNA is targeted to a sequence selected from SEQ ID 319 to SEQ ID 374.
25. The use of claim 17 wherein the target gene is prostaglandin endoperoxide synthase 2, and the siNA is targeted to a sequence selected from SEQ ID 375 to SEQ ID 491.
26. The use of claim 17 wherein the target gene is carbonic anhydrase XII, and the siNA is targeted to a sequence selected from SEQ ID 492 to SEQ ID 538.
27. The use of claim 17 wherein the target gene is alpha adrenergic receptor 1A, and the siNA is targeted to a sequence selected from SEQ ID 539 to SEQ ID 598.
28. The use of claim 17 wherein the target gene is alpha adrenergic receptor 1B, and the siNA is targeted to a sequence selected from SEQ ID 599 to SEQ ID 634.
29. The use of claim 17 wherein the target gene is alpha adrenergic receptor 1D, and the siNA is targeted to a sequence selected from SEQ ID 635 to SEQ ID 646.
30. The use of claim 17 wherein the target gene is angiotensinogen, and the siNA is targeted to a sequence selected from SEQ ID 647 to SEQ ID 694.
31. The use of claim 17 wherein the target gene is angiotensin II receptor type 1, and the siNA is targeted to a sequence selected from SEQ ID 695 to SEQ ID 749.
32. The use of claim 17 wherein the target gene is angiotensin II receptor type 2, and the siNA is targeted to a sequence selected from SEQ ID 750 to SEQ ID 807.
33. The use of claim 17 wherein the target gene is angiotensin I converting enzyme 1, and the siNA is targeted to a sequence selected from SEQ ID 808 to SEQ ID 939.
34. The use of claim 17 wherein the target gene is angiotensin I converting enzyme 2, and the siNA is targeted to a sequence selected from SEQ ID 940 to SEQ ID 1139.
35. The use of claim 17 wherein the target gene is renin, and the siNA is targeted to a sequence selected from SEQ ID 1140 to SEQ ID 1196.
36. The use of claim 17 wherein the target gene is cochlin, and the siNA is targeted to a sequence selected from SEQ ID 1197 to SEQ ID 1307.
37. The use of claim 17 wherein the target gene is ATPase alpha 1, and the siNA is targeted to a sequence selected from SEQ ID 1308 to SEQ ID 1500.
38. The use of claim 17 wherein the target gene is ATPase alpha 2, and the siNA is targeted to a sequence selected from SEQ ID 1501 to SEQ ID 1606.
39. The use of claim 17 wherein the target gene is ATPase alpha 3, and the siNA is targeted to a sequence selected from SEQ ID 1607 to SEQ ID 1705.
40. The use of claim 17 wherein the target gene is ATPase beta 1, and the siNA is targeted to a sequence selected from SEQ ID 1706 to SEQ ID 1780.
41. The use of claim 17 wherein the target gene is ATPase beta 2, and the siNA is targeted to a sequence selected from SEQ ID 1780 to SEQ ID 1829.
42. A method of treatment of an eye condition, comprising administering siNA to downregulate expression of a target gene selected from the group comprising aqueous formation genes and aqueous outflow genes in a patient.
43. The use of claim 42, wherein the eye condition is selected from the group comprising glaucoma, infection, inflammation, uveitis, and expression of systemic diseases.
44. The method of claim 42, wherein the eye condition is glaucoma.
45. The method of claim 42, wherein the eye condition is diabetic retinopathy.
46. The method of claim 42, wherein the target gene is selected from the group comprising: Carbonic Anhydrases II, IV and XII; Adrenergic Receptors: beta1 and 2 and alpha 1A, 1Band 1D; Acetylcholinesterase; Cyclooxygenases 1 and 2; ATPases: alpha1, alpha2, alpha3, beta1, beta2; Endothelial Leukocyte Adhesion Molecule I (ELAM-1); Angiotensin System: Angiotensin II, Angiotensin II Converting Enzymes (ACE I and ACE II), Angiotensin II Receptors (ATR1 and ATR2) and Renin; Cochlin.
47. The method of claim 42 wherein the siNA is siRNA.
48. The method of claim 47 wherein the siRNA is dsRNA.
49. The method of claim 47 wherein the siRNA is shRNA,
50. The method of claim 42 wherein the siNA comprises a modified oligonucleotide.
51. An isolated siNA molecule for use in treatment of an eye condition characterised by an altered intra-ocular pressure (IOP) in the patient, the siNA being complementary to a nucleotide sequence selected from SEQ ID 1 to SEQ ID 1829.
52. Use of an Isolated siNA molecule having a sequence which is complementary to a nucleotide sequence selected from SEQ ID 1 to SEQ ID 1829 in the preparation of a medicament for the treatment of an eye condition.
53. A pharmaceutical composition comprising siNA having a sequence which is complementary to a nucleotide sequence selected from SEQ ID 1 to SEQ ID 1829.
US11/574,169 2004-08-23 2005-08-23 Treatment of eye disorders characterized by an elevated intraocular pressure by siRNAs Active US8030284B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB0418762.1A GB0418762D0 (en) 2004-08-23 2004-08-23 Methods and compositions to treat glaucoma
GB0418762.1 2004-08-23
GB0503412.9 2005-02-18
GB0503412A GB0503412D0 (en) 2005-02-18 2005-02-18 Methods and compositions to treat glaucoma
PCT/GB2005/050134 WO2006021817A2 (en) 2004-08-23 2005-08-23 Treatment of eye disorders characterized by an elevated introacular pressure by sirnas

Publications (2)

Publication Number Publication Date
US20070270365A1 true US20070270365A1 (en) 2007-11-22
US8030284B2 US8030284B2 (en) 2011-10-04

Family

ID=35967904

Family Applications (11)

Application Number Title Priority Date Filing Date
US11/574,169 Active US8030284B2 (en) 2004-08-23 2005-08-23 Treatment of eye disorders characterized by an elevated intraocular pressure by siRNAs
US11/360,305 Active 2025-11-27 US7592325B2 (en) 2004-08-23 2006-02-22 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,157 Active 2025-10-21 US8252759B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,104 Active 2025-10-25 US8247386B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,148 Active 2025-10-23 US8252758B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,078 Active 2025-09-09 US8198250B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,116 Active 2025-11-04 US8247387B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,132 Active 2025-11-07 US8258110B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/563,530 Active US7902169B2 (en) 2004-08-23 2009-09-21 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/874,928 Active 2026-01-04 US8389490B2 (en) 2004-08-23 2010-09-02 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US13/744,881 Active US8951982B2 (en) 2004-08-23 2013-01-18 Methods and compositions for the treatment of eye disorders with increased intraocular pressure

Family Applications After (10)

Application Number Title Priority Date Filing Date
US11/360,305 Active 2025-11-27 US7592325B2 (en) 2004-08-23 2006-02-22 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,157 Active 2025-10-21 US8252759B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,104 Active 2025-10-25 US8247386B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,148 Active 2025-10-23 US8252758B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,078 Active 2025-09-09 US8198250B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,116 Active 2025-11-04 US8247387B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/170,132 Active 2025-11-07 US8258110B2 (en) 2004-08-23 2008-07-09 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/563,530 Active US7902169B2 (en) 2004-08-23 2009-09-21 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US12/874,928 Active 2026-01-04 US8389490B2 (en) 2004-08-23 2010-09-02 Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US13/744,881 Active US8951982B2 (en) 2004-08-23 2013-01-18 Methods and compositions for the treatment of eye disorders with increased intraocular pressure

Country Status (17)

Country Link
US (11) US8030284B2 (en)
EP (3) EP2292757A3 (en)
JP (2) JP5766900B2 (en)
CN (2) CN102895673A (en)
AU (1) AU2005276245C1 (en)
CA (1) CA2578064C (en)
CY (1) CY1119061T1 (en)
DK (1) DK1781787T3 (en)
ES (1) ES2632913T3 (en)
HR (1) HRP20171010T8 (en)
HU (1) HUE033977T2 (en)
LT (1) LT1781787T (en)
PL (1) PL1781787T3 (en)
PT (1) PT1781787T (en)
RU (1) RU2407531C2 (en)
SI (1) SI1781787T1 (en)
WO (1) WO2006021817A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140171487A1 (en) * 2005-12-28 2014-06-19 Alcon Research, Ltd. RNAi-MEDIATED INHIBITION OF PHOSPHODIESTERASE TYPE 4 FOR TREATMENT OF cAMP-RELATED OCULAR DISORDERS
US20160237440A1 (en) * 2013-10-22 2016-08-18 Sylentis Sau siRNA AND THEIR USE IN METHODS AND COMPOSITIONS FOR INHIBITING THE EXPRESSION OF THE PDK1 GENE
US10934542B2 (en) 2013-12-27 2021-03-02 Bonac Corporation Artificial match-type miRNA for controlling gene expression and use therefor
US11027023B2 (en) 2014-12-27 2021-06-08 Bonac Corporation Natural type miRNA for controlling gene expression, and use of same
US11142769B2 (en) 2015-03-27 2021-10-12 Bonac Corporation Single-stranded nucleic acid molecule having delivery function and gene expression regulating ability

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8076060B2 (en) * 2003-08-04 2011-12-13 Emil William Chynn Vaccination and immunotherapy as new therapeutic modalities in the treatment of glaucoma
ES2632913T3 (en) 2004-08-23 2017-09-18 Sylentis S.A.U. Treatment with mRNA of eye disorders characterized by elevated intraocular pressure
TW200639253A (en) * 2005-02-01 2006-11-16 Alcon Inc RNAi-mediated inhibition of ocular targets
WO2006094014A2 (en) 2005-02-28 2006-09-08 The Regents Of The University Of California Methods for diagnosis and treatment of endometrial cancer
WO2006113526A2 (en) 2005-04-15 2006-10-26 The Regents Of The University Of California Prevention of chlamydia infection using a protective antibody
US8318906B2 (en) 2005-04-15 2012-11-27 The Regents Of The University Of California EMP2 antibodies and their therapeutic uses
US8648052B2 (en) * 2005-04-15 2014-02-11 The Regents Of The University Of California Prevention of chlamydia infection using SIRNA
GB0521351D0 (en) * 2005-10-20 2005-11-30 Genomica Sau Modulation of TRPV expression levels
GB0521716D0 (en) * 2005-10-25 2005-11-30 Genomica Sau Modulation of 11beta-hydroxysteriod dehydrogenase 1 expression for the treatment of ocular diseases
CA2674210A1 (en) * 2005-12-30 2007-07-12 Intradigm Corporation Sirna compositions promoting scar-free wound healing of skin and methods for wound treatment
EP1977246B1 (en) * 2005-12-30 2010-01-20 Ventana Medical Systems, Inc. Na+, k+-atpase expression in cervical dysplasia and cancer
EP2423332A1 (en) 2006-08-25 2012-02-29 Oncotherapy Science, Inc. Prognostic markers and therapeutic targets for lung cancer
JP2010509251A (en) * 2006-11-09 2010-03-25 ユニバイオスクリーン エス.アー. Targeting alpha-1 or alpha-3 subunits of Na +, K + -ATPase in the treatment of proliferative diseases
JP2010512730A (en) 2006-12-13 2010-04-30 オンコセラピー・サイエンス株式会社 TTK as a tumor marker and therapeutic target for lung cancer
BRPI0815769A2 (en) 2007-08-24 2017-08-08 Oncotherapy Science Inc EBI3, DLX5, NPTX1 AND CDKN3 AS TARGET GENES FOR THE THERAPY AND DIAGNOSIS OF LUNG CANCER
BRPI0816150A2 (en) 2007-08-24 2019-09-24 Oncotherapy Science Inc cancer related genes, cdca5, eph47, stk31 and w-dhd1.
WO2011063161A2 (en) 2009-11-20 2011-05-26 The Regents Of The University Of California Epithelial membrane protein-2 (emp2) and proliferative vitreoretinopathy (pvr)
EP2329044B1 (en) 2008-08-27 2016-05-18 Oncotherapy Science, Inc. Prmt1 for target genes of cancer therapy and diagnosis
CN108165548B (en) 2008-09-22 2022-10-14 菲奥医药公司 Reduced size self-delivering RNAi compounds
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
KR20180044433A (en) 2010-03-24 2018-05-02 알엑스아이 파마슈티칼스 코포레이션 Rna interference in dermal and fibrotic indications
EP2550001B1 (en) 2010-03-24 2019-05-22 Phio Pharmaceuticals Corp. Rna interference in ocular indications
WO2012021107A2 (en) 2010-08-12 2012-02-16 Nanyang Technological University A liposomal formulation for ocular drug delivery
EP2638160B8 (en) 2010-11-10 2017-08-02 Nigel L. Webb Nuclions and ribocapsids
RU2460517C1 (en) * 2011-07-14 2012-09-10 Федеральное государственное учреждение "Микрохирургия глаза" имени академика С.Н. Федорова Федерального агентства по высокотехнологичной медицинской помощи" Pharmaceutical composition for integrated treatment of ocular surface diseases in patients suffering primary open-angle glaucoma
WO2014037686A1 (en) * 2012-09-05 2014-03-13 Sylentis S.A.U. Sirna and their use in methods and compositions for the treatment and/or prevention of eye conditions
GB201215857D0 (en) 2012-09-05 2012-10-24 Sylentis Sau siRNA and their use in methods and compositions for the treatment and/or prevention of eye conditions
CN105143203A (en) 2013-04-17 2015-12-09 辉瑞大药厂 N-piperidin-3-ylbenzamide derivatives for treating cardiovascular diseases
US8908303B1 (en) 2013-07-30 2014-12-09 Kabushiki Kaisha Toshiba Method for servo demodulation and disk storage apparatus
EP2865758A1 (en) * 2013-10-22 2015-04-29 Sylentis, S.A.U. siRNA and their use in methods and compositions for inhibiting the expression of the ORAI1 gene
US9956195B2 (en) 2014-01-07 2018-05-01 Nanyang Technological University Stable liposomal formulations for ocular drug delivery
US10011837B2 (en) 2014-03-04 2018-07-03 Sylentis Sau SiRNAs and their use in methods and compositions for the treatment and/or prevention of eye conditions
EP3137119B1 (en) 2014-04-28 2020-07-01 Phio Pharmaceuticals Corp. Methods for treating cancer using a nucleic acid targeting mdm2
WO2015168605A1 (en) * 2014-05-01 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treatment of disorders in the front of the eye utilizing nucleic acid molecules
FR3031503A1 (en) * 2015-01-14 2016-07-15 Qualipac Sa CONTAINER, COMPONENT OF CONTAINER, AND RANGE OF SUCH PRODUCTS
CA3009769C (en) * 2015-12-29 2021-01-12 National University Corporation Hokkaido University Single-stranded nucleic acid molecule inhibiting expression of prorenin gene or prorenin receptor gene, and use thereof
US10799676B2 (en) 2016-03-21 2020-10-13 Edwards Lifesciences Corporation Multi-direction steerable handles for steering catheters
CN106267222B (en) * 2016-09-12 2020-05-19 北京市心肺血管疾病研究所 Use of angiotensin ii for improving cardiac delivery of macromolecular drugs or drug carriers
CN106228753A (en) * 2016-09-30 2016-12-14 福建工程学院 A kind of accidental falls monitoring method, system and terminal
JP2020509752A (en) * 2017-03-10 2020-04-02 ザ メディカル カレッジ オブ ウィスコンシン インクThe Medical College Of Wisconsin, Inc. Riboswitch-regulated gene therapy for retinal diseases
JP2017200928A (en) * 2017-05-31 2017-11-09 シレンティス・エセ・ア・ウ Sirna and use thereof in method and composition for treatment and/or prevention of eye conditions
BR112021009787A2 (en) * 2018-11-29 2021-08-17 Ojai Retinal Technology, Llc process to deliver retinal phototherapy safely
AU2020210630A1 (en) 2019-01-23 2021-08-05 Regeneron Pharmaceuticals, Inc. Treatment of ophthalmic conditions with angiopoietin-like 7 (ANGPTL7) inhibitors
US11845989B2 (en) 2019-01-23 2023-12-19 Regeneron Pharmaceuticals, Inc. Treatment of ophthalmic conditions with angiopoietin-like 7 (ANGPTL7) inhibitors
US20210348234A1 (en) * 2020-02-24 2021-11-11 The Board Of Regents Of The University Of Texas System Molecular biomarkers and targets for fuches' endothelial corneal dystrophy and glaucoma
WO2021206917A1 (en) * 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021255262A1 (en) 2020-06-19 2021-12-23 Sylentis Sau siRNA AND COMPOSITIONS FOR PROPHYLACTIC AND THERAPEUTIC TREATMENT OF VIRUS DISEASES
KR20230148819A (en) 2021-02-26 2023-10-25 리제너론 파마슈티칼스 인코포레이티드 Treatment of inflammation with glucocorticoids and angiopoietin-like 7 (ANGPTL7) inhibitors

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4617299A (en) * 1983-12-19 1986-10-14 Knepper Paul A Method for the prevention of ocular hypertension, treatment of glaucoma and treatment of ocular hypertension
US4652586A (en) * 1980-11-07 1987-03-24 The General Hospital Corporation Selective beta-2 adrenergic antagonists for the treatment of glaucoma
US4757089A (en) * 1985-06-14 1988-07-12 Massachusetts Eye And Ear Infirmary Increasing aqueous humor outflow
US4812448A (en) * 1984-10-22 1989-03-14 Knepper Paul A Method for the prevention of ocular hypertension, treatment of glaucoma and treatment of ocular hypertension
US5075323A (en) * 1988-08-24 1991-12-24 Aktiebolaget Hassle Compounds including omeprazole in the treatment of glaucoma
US5260059A (en) * 1989-04-14 1993-11-09 The State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of Oregon Health Sciences University Treatment of open-angle glaucoma by modulation matrix metalloproteinases and their inhibitor
US5464866A (en) * 1992-08-17 1995-11-07 Alcon Laboratories, Inc. Substituted hydrindanes for the treatment of angiogenesis-dependent diseases
US5545626A (en) * 1994-01-19 1996-08-13 The Trustees Of Columbia University In The City Of New York Method of treating glaucoma with oligonucleotides
US5585401A (en) * 1994-12-09 1996-12-17 The Reents Of The University Of California Method for enhancing outflow of aqueous humor in treatment of glaucoma
US6365576B1 (en) * 1999-06-24 2002-04-02 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Method for inhibiting herpes infection
US6372249B1 (en) * 1991-12-16 2002-04-16 Baylor College Of Medicine Senscent cell-derived inhibitors of DNA synthesis
US20020165158A1 (en) * 2001-03-27 2002-11-07 King George L. Methods of modulating angiogenesis
US20040115641A1 (en) * 2002-12-11 2004-06-17 Isis Pharmaceuticals Inc. Modulation of ROCK 1 expression
US20040209832A1 (en) * 2001-11-30 2004-10-21 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20040235031A1 (en) * 2003-05-01 2004-11-25 Schultz Gregory S. Anti-scarring ribozymes and methods
US20050171039A1 (en) * 2002-02-20 2005-08-04 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20060094032A1 (en) * 2004-03-12 2006-05-04 Fougerolles Antonin D iRNA agents targeting VEGF
US20060172965A1 (en) * 2005-02-01 2006-08-03 Alcon, Inc. RNAi-mediated inhibition of ocular targets
US20070093435A1 (en) * 2003-03-28 2007-04-26 Peter Andrews Modulation of cell phenotype by inhibitory rna
US20070167384A1 (en) * 2003-04-02 2007-07-19 Dharmacon, Inc. Modified polynucleotides for use in rna interference
US7521431B2 (en) * 2002-11-01 2009-04-21 The Trustees Of The University Of Pennsylvania Compositions and methods for siRNA inhibition of HIF-1 alpha
US7592325B2 (en) * 2004-08-23 2009-09-22 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure

Family Cites Families (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4343794A (en) 1980-05-06 1982-08-10 Mt. Sinai Method of reducing intraocular pressure with salts of vanadic acid
US5242943A (en) 1984-03-28 1993-09-07 William J. Louis 3-aminopropoxyphenyl derivatives, their preparation and pharmaceutical compositions containing them
US20020055536A1 (en) 1996-09-26 2002-05-09 Dewitte Robert S. System and method for structure-based drug design that includes accurate prediction of binding free energy
US6489307B1 (en) 1998-09-14 2002-12-03 University Of Florida Antisense compositions targeted to β1-adrenoceptor-specific mRNA and methods of use
AU776150B2 (en) 1999-01-28 2004-08-26 Medical College Of Georgia Research Institute, Inc. Composition and method for (in vivo) and (in vitro) attenuation of gene expression using double stranded RNA
US20040259247A1 (en) 2000-12-01 2004-12-23 Thomas Tuschl Rna interference mediating small rna molecules
US20070049543A1 (en) * 2001-05-18 2007-03-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of 11 beta-hydroxysteroid dehydrogenase-1 (11 beta-HSD-1) gene expression using short interfering nucleic acid siNA
EP1587574A4 (en) 2001-12-21 2009-03-18 Rhode Island Hospital SELECTIVE 11b-HSD INHIBITORS AND METHODS FOR USE THEREOF
US7294504B1 (en) 2001-12-27 2007-11-13 Allele Biotechnology & Pharmaceuticals, Inc. Methods and compositions for DNA mediated gene silencing
GB2406568B (en) 2002-02-20 2005-09-28 Sirna Therapeutics Inc RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
EP1432724A4 (en) 2002-02-20 2006-02-01 Sirna Therapeutics Inc Rna interference mediated inhibition of map kinase genes
US20050222061A1 (en) * 2002-04-18 2005-10-06 Schulte Ralf W Means and methods for the specific inhibition of genes in cells and tissue of the cns and/or eye
JP2005523928A (en) 2002-04-30 2005-08-11 アルコン,インコーポレイテッド Modulates, inhibits, or modulates the activity and / or expression of connective tissue growth factor (CTGF) as a unique means for both reducing intraocular pressure and treating glaucomatous retinopathy / optic neuropathy Drugs
US20040146858A1 (en) 2002-07-24 2004-07-29 Immusol, Inc. Novel siRNA gene libraries and methods for their production and use
US20040115815A1 (en) 2002-07-24 2004-06-17 Immusol, Inc. Single promoter system for making siRNA expression cassettes and expression libraries using a polymerase primer hairpin linker
EP1857547B2 (en) 2002-08-05 2020-12-02 Silence Therapeutics GmbH Further novel forms of interfering RNA molecules
US20040029275A1 (en) 2002-08-10 2004-02-12 David Brown Methods and compositions for reducing target gene expression using cocktails of siRNAs or constructs expressing siRNAs
EP1556402B1 (en) 2002-09-25 2011-06-22 University of Massachusetts In vivo gene silencing by chemically modified and stable sirna
DE10322662A1 (en) 2002-11-06 2004-10-07 Grünenthal GmbH New DNA type 10-23 enzyme, useful for treating e.g. pain, and related short interfering RNA, directed against the vanillin receptor or picorna viruses, contains specific nucleotide modifications for improved stability
CN1498964A (en) 2002-11-07 2004-05-26 本元正阳基因技术股份有限公司 Serial recombined gland related virus inducible path of RNAi, and utilized in gene therapy
US7250496B2 (en) * 2002-11-14 2007-07-31 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory genes and uses thereof
ES2440284T3 (en) * 2002-11-14 2014-01-28 Thermo Fisher Scientific Biosciences Inc. SiRNA directed to tp53
WO2006006948A2 (en) 2002-11-14 2006-01-19 Dharmacon, Inc. METHODS AND COMPOSITIONS FOR SELECTING siRNA OF IMPROVED FUNCTIONALITY
US20060257851A1 (en) * 2002-11-26 2006-11-16 Itzhak Bentwich Bioinformatically detectable group of novel viral regulatory genes and uses thereof
US20040167090A1 (en) 2003-02-21 2004-08-26 Monahan Sean D. Covalent modification of RNA for in vitro and in vivo delivery
US20040198640A1 (en) 2003-04-02 2004-10-07 Dharmacon, Inc. Stabilized polynucleotides for use in RNA interference
US20040224405A1 (en) 2003-05-06 2004-11-11 Dharmacon Inc. siRNA induced systemic gene silencing in mammalian systems
EP1636342A4 (en) 2003-06-20 2008-10-08 Isis Pharmaceuticals Inc Oligomeric compounds for use in gene modulation
US20050197364A1 (en) 2003-10-07 2005-09-08 Kelly Michael G. Amide compounds as ion channel ligands and uses thereof
GB0324334D0 (en) 2003-10-17 2003-11-19 Pilkington Plc Load bearing laminates
WO2005045037A2 (en) 2003-10-23 2005-05-19 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF 5-ALPHA REDUCTASE AND ANDROGEN RECEPTOR GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20050208658A1 (en) * 2003-11-21 2005-09-22 The University Of Maryland RNA interference mediated inhibition of 11beta hydroxysteriod dehydrogenase-1 (11beta HSD-1) gene expression
WO2005076998A2 (en) * 2004-02-05 2005-08-25 Intradigm Corporation Rnai therapeutics for treatment of eye neovascularization diseases
GB0403600D0 (en) * 2004-02-18 2004-03-24 Trinity College Dublin Methods and reagents for treating disease
WO2005117938A2 (en) 2004-04-13 2005-12-15 Regents Of The University Of Minnesota Methods of treating ocular conditions
CA2566519C (en) 2004-05-14 2020-04-21 Rosetta Genomics Ltd. Micrornas and uses thereof
US7687665B2 (en) * 2004-06-24 2010-03-30 Incyte Corporation 2-methylprop anamides and their use as pharmaceuticals
AU2005272816B2 (en) 2004-08-10 2011-08-11 Alnylam Pharmaceuticals, Inc. Chemically modified oligonucleotides
GB0421169D0 (en) 2004-09-23 2004-10-27 Securis Ltd Apparatus and process for the printing of microstructures
JP2008533050A (en) 2005-03-11 2008-08-21 アルコン,インコーポレイテッド RNAI-mediated inhibition of Frizzled-related protein-1 to treat glaucoma
US7621431B2 (en) * 2005-04-06 2009-11-24 Chicco Usa, Inc. Folding stroller tray
KR101283539B1 (en) * 2007-08-29 2013-07-15 삼성전자주식회사 Inverted non-volatile memory devices, stack modules and method of fabricating the same

Patent Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4652586A (en) * 1980-11-07 1987-03-24 The General Hospital Corporation Selective beta-2 adrenergic antagonists for the treatment of glaucoma
US4617299A (en) * 1983-12-19 1986-10-14 Knepper Paul A Method for the prevention of ocular hypertension, treatment of glaucoma and treatment of ocular hypertension
US4812448A (en) * 1984-10-22 1989-03-14 Knepper Paul A Method for the prevention of ocular hypertension, treatment of glaucoma and treatment of ocular hypertension
US4757089A (en) * 1985-06-14 1988-07-12 Massachusetts Eye And Ear Infirmary Increasing aqueous humor outflow
US5075323A (en) * 1988-08-24 1991-12-24 Aktiebolaget Hassle Compounds including omeprazole in the treatment of glaucoma
US5260059A (en) * 1989-04-14 1993-11-09 The State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of Oregon Health Sciences University Treatment of open-angle glaucoma by modulation matrix metalloproteinases and their inhibitor
US6372249B1 (en) * 1991-12-16 2002-04-16 Baylor College Of Medicine Senscent cell-derived inhibitors of DNA synthesis
US5464866A (en) * 1992-08-17 1995-11-07 Alcon Laboratories, Inc. Substituted hydrindanes for the treatment of angiogenesis-dependent diseases
US5545626A (en) * 1994-01-19 1996-08-13 The Trustees Of Columbia University In The City Of New York Method of treating glaucoma with oligonucleotides
US5585401A (en) * 1994-12-09 1996-12-17 The Reents Of The University Of California Method for enhancing outflow of aqueous humor in treatment of glaucoma
US6365576B1 (en) * 1999-06-24 2002-04-02 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Method for inhibiting herpes infection
US20020165158A1 (en) * 2001-03-27 2002-11-07 King George L. Methods of modulating angiogenesis
US20040209832A1 (en) * 2001-11-30 2004-10-21 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US7176304B2 (en) * 2002-02-20 2007-02-13 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20050171039A1 (en) * 2002-02-20 2005-08-04 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US7521431B2 (en) * 2002-11-01 2009-04-21 The Trustees Of The University Of Pennsylvania Compositions and methods for siRNA inhibition of HIF-1 alpha
US20040115641A1 (en) * 2002-12-11 2004-06-17 Isis Pharmaceuticals Inc. Modulation of ROCK 1 expression
US20070093435A1 (en) * 2003-03-28 2007-04-26 Peter Andrews Modulation of cell phenotype by inhibitory rna
US20070167384A1 (en) * 2003-04-02 2007-07-19 Dharmacon, Inc. Modified polynucleotides for use in rna interference
US7462602B2 (en) * 2003-05-01 2008-12-09 University Of Florida Research Foundation, Inc. Anti-scarring ribozymes and methods
US20040235031A1 (en) * 2003-05-01 2004-11-25 Schultz Gregory S. Anti-scarring ribozymes and methods
US20060094032A1 (en) * 2004-03-12 2006-05-04 Fougerolles Antonin D iRNA agents targeting VEGF
US7592325B2 (en) * 2004-08-23 2009-09-22 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US20060172965A1 (en) * 2005-02-01 2006-08-03 Alcon, Inc. RNAi-mediated inhibition of ocular targets
US20060172963A1 (en) * 2005-02-01 2006-08-03 Alcon, Inc. RNAi-mediated inhibition of ocular hypertension targets
US7592324B2 (en) * 2005-02-01 2009-09-22 Alcon, Inc. RNAi-mediated inhibition of ocular targets
US20090326044A1 (en) * 2005-02-01 2009-12-31 Alcon Research, Ltd. RNAi-Mediated Inhibition of Ocular Targets

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140171487A1 (en) * 2005-12-28 2014-06-19 Alcon Research, Ltd. RNAi-MEDIATED INHIBITION OF PHOSPHODIESTERASE TYPE 4 FOR TREATMENT OF cAMP-RELATED OCULAR DISORDERS
US9453220B2 (en) * 2005-12-28 2016-09-27 Arrowhead Pharmaceuticals, Inc. RNAi-mediated inhibition of phosphodiesterase type 4 for treatment of cAMP-related ocular disorders
US9765340B2 (en) * 2005-12-28 2017-09-19 Arrowhead Pharmaceuticals, Inc. RNAi-mediated inhibition of phosphodiesterase type 4 for treatment of CAMP-related ocular disorders
US20160237440A1 (en) * 2013-10-22 2016-08-18 Sylentis Sau siRNA AND THEIR USE IN METHODS AND COMPOSITIONS FOR INHIBITING THE EXPRESSION OF THE PDK1 GENE
US9951338B2 (en) * 2013-10-22 2018-04-24 Sylentis Sau siRNA and their use in methods and compositions for inhibiting the expression of the PDK1 gene
US10934542B2 (en) 2013-12-27 2021-03-02 Bonac Corporation Artificial match-type miRNA for controlling gene expression and use therefor
US11027023B2 (en) 2014-12-27 2021-06-08 Bonac Corporation Natural type miRNA for controlling gene expression, and use of same
US11142769B2 (en) 2015-03-27 2021-10-12 Bonac Corporation Single-stranded nucleic acid molecule having delivery function and gene expression regulating ability

Also Published As

Publication number Publication date
US20060234970A1 (en) 2006-10-19
DK1781787T3 (en) 2017-07-31
AU2005276245A1 (en) 2006-03-02
US8247386B2 (en) 2012-08-21
CY1119061T1 (en) 2018-01-10
SI1781787T1 (en) 2017-08-31
US8198250B2 (en) 2012-06-12
CA2578064A1 (en) 2006-03-02
JP2012205594A (en) 2012-10-25
AU2005276245B2 (en) 2010-06-03
PT1781787T (en) 2017-07-17
US20090099112A1 (en) 2009-04-16
CN102895673A (en) 2013-01-30
WO2006021817A3 (en) 2006-06-08
HRP20171010T8 (en) 2017-11-03
ES2632913T3 (en) 2017-09-18
RU2007110646A (en) 2008-09-27
US8389490B2 (en) 2013-03-05
US8030284B2 (en) 2011-10-04
US8252758B2 (en) 2012-08-28
US8247387B2 (en) 2012-08-21
JP2008510786A (en) 2008-04-10
PL1781787T3 (en) 2018-01-31
AU2005276245C1 (en) 2015-02-26
JP5766900B2 (en) 2015-08-19
HUE033977T2 (en) 2018-02-28
US20140018527A1 (en) 2014-01-16
CN105251024A (en) 2016-01-20
US20100022623A1 (en) 2010-01-28
US20100331394A1 (en) 2010-12-30
US20090099113A1 (en) 2009-04-16
RU2407531C2 (en) 2010-12-27
EP2292757A2 (en) 2011-03-09
US8252759B2 (en) 2012-08-28
EP1781787B1 (en) 2017-04-12
US20090099111A1 (en) 2009-04-16
EP1781787A2 (en) 2007-05-09
EP2298892A2 (en) 2011-03-23
US7902169B2 (en) 2011-03-08
EP2298892A3 (en) 2011-11-16
HRP20171010T1 (en) 2017-09-22
LT1781787T (en) 2017-07-25
WO2006021817A2 (en) 2006-03-02
EP2292757A3 (en) 2011-11-16
US8258110B2 (en) 2012-09-04
US20090105175A1 (en) 2009-04-23
US20090099114A1 (en) 2009-04-16
CA2578064C (en) 2015-10-13
US8951982B2 (en) 2015-02-10
US20090105176A1 (en) 2009-04-23
US7592325B2 (en) 2009-09-22

Similar Documents

Publication Publication Date Title
US8030284B2 (en) Treatment of eye disorders characterized by an elevated intraocular pressure by siRNAs
EP1941037B1 (en) Modulation of 11beta-hydroxysteroid dehydrogenase 1 expression for the treatment of ocular diseases

Legal Events

Date Code Title Description
AS Assignment

Owner name: SYLENTIS S.A.U., SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JIMENEZ, ANA I.;SESTO, ANGELA;ROMAN, JOSE P.;AND OTHERS;REEL/FRAME:019549/0026;SIGNING DATES FROM 20070601 TO 20070628

Owner name: SYLENTIS S.A.U., SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JIMENEZ, ANA I.;SESTO, ANGELA;ROMAN, JOSE P.;AND OTHERS;SIGNING DATES FROM 20070601 TO 20070628;REEL/FRAME:019549/0026

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1553); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 12